Phosphatidylinositol 3-kinase inhibitors

- Gilead Sciences, Inc.

The present application provides the compounds of formula (J), or pharmaceutically acceptable salts, isomers, tautomer, or a mixture thereof, wherein n, W, A′, B′, R1, R2, and R3 are described herein. The compounds are inhibitors to the activities of phosphatidylinositol 3-kinase (PI3K) and are useful for treating conditions mediated by one or more PI3K isoforms. The present application further provides pharmaceutical compositions that include a compound of formula (I), or pharmaceutically acceptable salts, isomers, tautomer, or mixture thereof, and methods of using these compounds and compositions for treating conditions mediated by one or more PI3K isoforms.

Skip to: Description  ·  Claims  ·  References Cited  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority to and the benefit under 35 U.S.C. 119(e) of U.S. Provisional Patent Application Ser. No. 62/012,172, filed Jun. 13, 2014, the disclosure of which is hereby incorporated by reference in its entirety.

FIELD

The present application relates to novel compounds that selectively inhibit the activities of PI3K isoforms and their uses in therapeutic treatments.

BACKGROUND

Cell signaling via 3′-phosphorylated phosphoinositides has been implicated in a variety of cellular processes, e.g., malignant transformation, growth factor signaling, inflammation, and immunity (Rameh et al., J. Biol. Chem., 274:8347-8350, 1999). Phosphatidylinositol 3-kinase (PI 3-kinase or PI3K) is responsible for generating these phosphorylated signaling products. PI3K was initially identified as a protein associated with viral oncoproteins and growth factor receptor tyrosine kinases that phosphorylate phosphatidylinositol (PI) and its phosphorylated derivatives at the 3′-hydroxyl of the inositol ring (Panayotou et al., Trends Cell Biol., 2:358-60, 1992).

Three classes of the PI 3-kinase (PI3K) are proposed based on the substrate specificities. Class I PI3Ks phosphorylate phosphatidylinositol (PI), phosphatidylinositol-4-phosphate, and phosphatidylinositol-4,5-biphosphate (PIP2) to produce phosphatidylinositol-3-phosphate (PIP), phosphatidylinositol-3,4-biphosphate, and phosphatidylinositol-3,4,5-triphosphate, respectively. Also, Class II PI3Ks phosphorylate PI and phosphatidylinositol-4-phosphate, and Class III PI3Ks phosphorylate PI.

The initial purification and molecular cloning of PI 3-kinase revealed that it was a heterodimer consisting of p85 and p110 subunits (Otsu et al., Cell, 65:91-104, 1991; Hiles et al., Cell, 70:419-29, 1992). Later, four distinct Class I PI3Ks were identified and designated as PI3Kα, β, δ, and γ isoforms. Each isoform consists of a distinct 110 kDa catalytic subunit and a regulatory subunit. The catalytic subunits of PI3Kα, β, and δ (i.e., p110α, p110β, and p110δ, respectively) interacts, individually, with the same regulatory subunit p85, whereas the catalytic subunit of P131γ (p110γ) interacts with a distinct regulatory subunit p101.

Studies have also showed that each PI3K isoform has distinct expression pattern. For example, PIK3CA which encodes PI3Kα is frequently mutated in human cancers (Engelman, Nat. Rev. Cancer, 9: 550-562, 2009). Also, PI3Kδ is generally expressed in hematopoietic cells. Moreover, PI3K isoforms are shown to be associated with proliferation or survival signaling in cancers, inflammatory, or autoimmune diseases. As each PI3K isoform has different biological function, PI3K isoforms are potential targets to treat cancer or disorder (U.S. Pat. Nos. 6,800,620; 8,435,988; 8,673,906; US Patent Application Publication No. US2013/0274253).

Therefore, there is a need for developing therapeutic agents that inhibit PI3K isoforms to treat diseases, disorders, or conditions that are mediated by PI3K.

SUMMARY

The present application provides novel compounds that are inhibitors of PI3K isoforms. The application also provides compositions, including pharmaceutical compositions, kits that include the compounds, and methods of using and making the compounds. The compounds provided herein are useful in treating diseases, disorders, or conditions that are mediated by PI3K isoforms. The application also provides compounds for use in therapy. The application further provides compounds for use in a method of treating a disease, disorder, or condition that is mediated by PI3K isoforms. Moreover, the application provides uses of the compounds in the manufacture of a medicament for the treatment of a disease, disorder or condition that is mediated by PI3K isoforms.

The applications provides the compounds having the structure of formula (I):

n is 0, 1, 2, 3, or 4;

m is 0 or 1;

A is a single bond or C(O);

B is alkyl or cycloalkyl, wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl or alkoxy;

R′ is hydrogen or optionally substituted alkyl;

R1 is independently selected from halo, cyano, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkoxy, optionally substituted sulfonyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;

R2 is cyano, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z; wherein R2x is hydrogen, optionally substituted alkyl, heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl; wherein R2y is hydrogen and alkyl, wherein R2z is alkyl;

R3 is hydrogen, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted cycloalkyl, or optionally substituted aryl;

R4 is heteroaryl optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted alkyl, and —NH2; and

R5 is hydrogen or optionally substituted alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The application also provides the compound having the structure of formula (I), wherein

n is 0, 1, 2 or 3;

m is 0 or 1;

A is a single bond or C(O);

B is C1-6 alkyl or C3-8 cycloalkyl, wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl or C1-6 alkoxy;

R′ is hydrogen or optionally substituted C1-6 alkyl;

each R1 is independently selected from halo, cyano, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted sulfonyl, optionally substituted C3-8 aryl, optionally substituted C3-8 heteroaryl, optionally substituted C3-8 cycloalkyl, and optionally substituted C2-8 heterocycloalkyl;

R2 is cyano, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z,

    • where Rx is hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, C4-8 heteroaryl;
    • wherein R2 is independently selected from hydrogen and C1-6 alkyl; and
    • wherein R2z is C1-6 alkyl;

R3 is hydrogen, optionally substituted C1-6 alkyl, C3-8 cycloalkyl, or C6-10 aryl;

R4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two heteroatoms selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted C1-6 alkyl, and —NH2; and

R5 is hydrogen or optionally substituted C1-6 alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a four- or eight-membered heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In some embodiments, the compounds have the structure of formula (I), wherein B is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, each of which is optionally substituted with methoxy, ethoxy, or hydroxyl. In certain embodiments, wherein R′ is methyl, ethyl, propyl, phenylmethyl, phenylethyl, or phenylpropyl. In other embodiments, wherein each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl. In some other embodiments, wherein R2 is selected from cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, or —SO2R2z; wherein each R2x is independently methyl, butyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl, oxetanyl, or morpholinyl, each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, or iodo; wherein each R2 is independently hydrogen, methyl, ethyl, propyl, butyl; and wherein each R2z is independently methyl, ethyl, propyl, or butyl. In certain other embodiments, wherein R3 is selected from hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, or phenyl. In additional embodiments, wherein R5 is hydrogen, methyl, ethyl, or propyl. In other additional embodiments, R5 and R3 together with the atoms to which they are attached optionally form pyrrolidinyl. In further embodiments, wherein R4 is a monocyclic heteroaryl having at least two nitrogen atoms, wherein R4 is optionally substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and —NH2. In yet further embodiments, wherein R4 is bicyclic heteroaryl having at least one aromatic ring, at least two nitrogen atoms, and at least one additional heteroatom selected from N, O, or S; wherein R4 is optionally substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and —NH2.

In certain embodiments, the PI3K inhibitors are the compounds selected from Table 1, a pharmaceutically acceptable salt, isomer, or a mixture thereof. In certain embodiments, the PI3K inhibitors are the compounds selected from Table 1a, a pharmaceutically acceptable salt, isomer, or a mixture thereof. In additional embodiments, the compound is an (S)-enantiomer. In other embodiments, the compound is an (R)-enantiomer. In other additional embodiments, the compound is an atropisomer.

The application also provides a pharmaceutical composition that comprises a compound of formula (I), a pharmaceutically acceptable salt, isomer, or a mixture thereof, together with at least one pharmaceutically acceptable vehicle. Examples of a pharmaceutically acceptable vehicle may be selected from carriers, adjuvants, and excipients.

Further provided herein is a method of treating a disease, disorder, or condition in a human in need thereof by administering to the human a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof. Further provided is a compound of formula (I) for use in a method of treating a disease, disorder or condition that is mediated by PI3K isoforms. The application also provides the use of a compound of formula (I) in the manufacture of a medicament for the treatment of a disease, disorder or condition that is mediated by PI3K isoforms. In certain embodiments, the disease, disorder, or condition is associated or mediated by PI3K. In some embodiments, the disease, disorder, or condition is an inflammatory disorder. In other embodiments, the disease, disorder, or condition is a cancer.

Also provided herein is a method of inhibiting the activity of a phosphatidylinositol 3-kinase polypeptide by contacting the polypeptide with a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

Further provided is a method of inhibiting excessive or destructive immune reactions, comprising administering an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

Also provided is a method of inhibiting growth or proliferation of cancer cells comprising contacting the cancer cells with an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

Also provided is a kit that includes a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof. The kit may further comprise a label and/or instructions for use of the compound in treating a disease, disorder, or condition in a human in need thereof. In some embodiments, the disease, disorder, or condition may be associated or mediated by PI3K activity.

Also provided are articles of manufacture that include a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof, and a container. In one embodiment, the container may be a vial, jar, ampoule, preloaded syringe, or an intravenous bag.

DETAILED DESCRIPTION

The following description sets forth exemplary methods, parameters and the like. Such description is not intended as a limitation on the scope of the present application but is instead provided as exemplary embodiments.

As used herein, the following words, phrases and symbols are generally intended to have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise.

A dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, —CONH2 is attached through the carbon atom. A dash at the front or end of a chemical group is a matter of convenience; chemical groups may be depicted with or without one or more dashes without losing their ordinary meaning. A wavy line drawn through a line in a structure indicates a point of attachment of a group. Unless chemically or structurally required, no directionality is indicated or implied by the order in which a chemical group is written or named.

The prefix “Cu-v” indicates that the following group has from u to v carbon atoms. For example, “C1-6 alkyl” indicates that the alkyl group has from 1 to 6 carbon atoms.

Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. In certain embodiments, the term “about” includes the indicated amount ±10%. In other embodiments, the term “about” includes the indicated amount ±5%. In certain other embodiments, the term “about” includes the indicated amount ±1%. Also, to the term “about X” includes description of “X”. Also, the singular forms “a” and “the” include plural references unless the context clearly dictates otherwise. Thus, e.g., reference to “the compound” includes a plurality of such compounds and reference to “the assay” includes reference to one or more assays and equivalents thereof known to those skilled in the art.

“Alkyl” refers to an unbranched or branched saturated hydrocarbon chain. As used herein, alkyl has 1 to 20 carbon atoms (i.e., C1-20 alkyl), 1 to 8 carbon atoms (i.e., C1-8 alkyl), 1 to 6 carbon atoms (i.e., C1-6 alkyl), or 1 to 4 carbon atoms (i.e., C1-4 alkyl). Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl, and 3-methylpentyl. When an alkyl residue having a specific number of carbons is named by chemical name or identified by molecular formula, all geometric isomers having that number of carbons may be encompassed; thus, for example, “butyl” includes n-butyl (i.e. —(CH2)3CH3), sec-butyl (i.e, —CH(CH3)CH2CH3), isobutyl (i.e. —CH2CH(CH3)2) and tert-butyl (i.e. —C(CH3)3); and “propyl” includes n-propyl (i.e. —(CH2)2CH3) and isopropyl (i.e. —CH(CH3)3).

“Alkenyl” refers to an aliphatic group containing at least one carbon-carbon double bond and having from 2 to 20 carbon atoms (i.e., C2-20 alkenyl), 2 to 8 carbon atoms (i.e., C2-8 alkenyl), 2 to 6 carbon atoms (i.e., C2-6 alkenyl), or 2 to 4 carbon atoms (i.e., C2-4 alkenyl). Examples of alkenyl groups include ethenyl, propenyl, butadienyl (including 1,2-butadienyl and 1,3-butadienyl).

“Alkynyl” refers to an aliphatic group containing at least one carbon-carbon triple bond and having from 2 to 20 carbon atoms (i.e., C2-20 alkynyl), 2 to 8 carbon atoms (i.e., C2-8 alkynyl), 2 to 6 carbon atoms (i.e., C2-6 alkynyl), or 2 to 4 carbon atoms (i.e., C2-4 alkynyl). The term “alkynyl” also includes those groups having one triple bond and one double bond.

“Alkoxy” refers to the group “alkyl-O—”. Examples of alkoxy groups include methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, and 1,2-dimethylbutoxy.

“Acyl” refers to a group —C(═O)R, wherein R is hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroalkyl, or heteroaryl; each of which may be optionally substituted, as defined herein. Examples of acyl include formyl, acetyl, cylcohexylcarbonyl, cyclohexylmethyl-carbonyl, and benzoyl.

“Amido” refers to both a “C-amido” group which refers to the group —C(═O)NRyRz and an “N-amido” group which refers to the group —NRyC(═O)Rz, wherein Ry and Rz are independently selected from the group consisting of hydrogen, alkyl, aryl, haloalkyl, or heteroaryl; each of which may be optionally substituted.

“Amino” refers to the group —NRyRz wherein Ry and Rz are independently selected from the group consisting of hydrogen, alkyl, haloalkyl, aryl, or heteroaryl; each of which may be optionally substituted.

“Aryl” refers to an aromatic carbocyclic group having a single ring (e.g. monocyclic) or multiple rings (e.g. bicyclic or tricyclic) including fused systems. As used herein, aryl has 6 to 20 ring carbon atoms (i.e., C6-20 aryl), 6 to 12 carbon ring atoms (i.e., C6-12 aryl), or 6 to 10 carbon ring atoms (i.e., C6-10 aryl). Examples of aryl groups include phenyl, naphthyl, fluorenyl, and anthryl. Aryl, however, does not encompass or overlap in any way with heteroaryl defined below. If one or more aryl groups are fused with a heteroaryl ring, the resulting ring system is heteroaryl.

“Cyano” or “carbonitrile” refers to the group —CN.

“Cycloalkyl” refers to a saturated or partially saturated cyclic alkyl group having a single ring or multiple rings including fused, bridged, and spiro ring systems. The term “cycloalkyl” includes cycloalkenyl groups (i.e. the cyclic group having at least one alkenyl). As used herein, cycloalkyl has from 3 to 20 ring carbon atoms (i.e., C3-20 cycloalkyl), 3 to 12 ring carbon atoms (i.e., C3-12 cycloalkyl), 3 to 10 ring carbon atoms (i.e., C3-10 cycloalkyl), 3 to 8 ring carbon atoms (i.e., C3-8 cycloalkyl), or 3 to 6 ring carbon atoms (i.e., C3-6 cycloalkyl). Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.

“Halogen” or “halo” includes fluoro, chloro, bromo, and iodo. “Haloalkyl” refers to an unbranched or branched alkyl group as defined above, wherein one or more hydrogen atoms are replaced by a halogen. For example, where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached. Dihaloalkyl and trihaloalkyl refer to alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be, but are not necessarily, the same halogen. Examples of haloalkyl include difluoromethyl (—CHF2) and trifluoromethyl (—CF3).

“Heteroalkyl” refers to an alkyl group in which one or more of the carbon atoms (and any associated hydrogen atoms) are each independently replaced with the same or different heteroatomic group. The term “heteroalkyl” includes unbranched or branched saturated chain having carbon and heteroatoms. By way of example, 1, 2 or 3 carbon atoms may be independently replaced with the same or different heteroatomic group. Heteroatomic groups include, but are not limited to, —NR—, —O—, —S—, —S(O)—, —S(O)2—, and the like, where R is H, alkyl, aryl, cycloalkyl, heteroalkyl, heteroaryl or heterocycloalkyl, each of which may be optionally substituted. Examples of heteroalkyl groups include —OCH3, —CH2OCH3, —SCH3, —CH2SCH3, —NRCH3, and —CH2NRCH3, where R is hydrogen, alkyl, aryl, arylalkyl, heteroalkyl, or heteroaryl, each of which may be optionally substituted. As used herein, heteroalkyl include 1 to 10 carbon atoms, 1 to 8 carbon atoms, or 1 to 4 carbon atoms; and 1 to 3 heteroatoms, 1 to 2 heteroatoms, or 1 heteroatom.

“Heteroaryl” refers to an aromatic group having a single ring, multiple rings, or multiple fused rings, with one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur. As used herein, heteroaryl include 1 to 20 ring carbon atoms (i.e., C1-20 heteroaryl), 3 to 12 ring carbon atoms (i.e., C3-12 heteroaryl), or 3 to 8 carbon ring atoms (i.e., C3-8 heteroaryl); and 1 to 5 heteroatoms, 1 to 4 heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups include pyrimidinyl, purinyl, pyridyl, pyridazinyl, benzothiazolyl, and pyrazolyl. Heteroaryl does not encompass or overlap with aryl as defined above.

“Heterocycloalkyl” refers to a saturated or unsaturated cyclic alkyl group, with one or more ring heteroatoms independently selected from nitrogen, oxygen and sulfur. The term “heterocycloalkyl” includes heterocycloalkenyl groups (i.e. the heterocycloalkyl group having at least one alkenyl). A heterocycloalkyl may be a single ring or multiple rings wherein the multiple rings may be fused, bridged, or spiro. As used herein, heterocycloalkyl has 2 to 20 ring carbon atoms (i.e., C2-20 heterocycloalkyl), 2 to 12 ring carbon atoms (i.e., C2-12 heterocycloalkyl), 2 to 10 ring carbon atoms (i.e., C2-10 heterocycloalkyl), 2 to 8 ring carbon atoms (i.e., C2-8 heterocycloalkyl), 3 to 12 ring carbon atoms (i.e., C3-12 heterocycloalkyl), 3 to 8 ring carbon atoms (i.e., C3-8 heterocycloalkyl), or 3 to 6 ring carbon atoms (i.e., C3-6 heterocycloalkyl); having 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, sulfur or oxygen. Examples of heterocycloalkyl groups include pyrrolidinyl, piperidinyl, piperazinyl, oxetanyl, dioxolanyl, azetidinyl, and morpholinyl.

“Hydroxy” or “hydroxyl” refers to the group —OH.

“Oxo” refers to the group (═O) or (O).

“Sulfonyl” refers to the group —S(O)2R, where R is alkyl, haloalkyl, heterocycloalkyl, cycloalkyl, heteroaryl, or aryl. Examples of sulfonyl are methylsulfonyl, ethylsulfonyl, phenylsulfonyl, and toluenesulfonyl.

Certain commonly used alternative chemical names may be used. For example, a divalent group such as a divalent “alkyl” group, a divalent “aryl” group, etc., may also be referred to as an “alkylene” group or an “alkylenyl” group, an “arylene” group or an “arylenyl” group, respectively. Also, unless indicated explicitly otherwise, where combinations of groups are referred to herein as one moiety, e.g. arylalkyl, the last mentioned group contains the atom by which the moiety is attached to the rest of the molecule.

The terms “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. Also, the term “optionally substituted” refers to any one or more hydrogen atoms on the designated atom or group may or may not be replaced by a moiety other than hydrogen.

The term “substituted” means that any one or more hydrogen atoms on the designated atom or group is replaced with one or more substituents other than hydrogen, provided that the designated atom's normal valence is not exceeded. The one or more substituents include, but are not limited to, alkyl, alkenyl, alkynyl, alkoxy, acyl, amino, amido, amidino, aryl, azido, carbamoyl, carboxyl, carboxyl ester, cyano, guanidino, halo, haloalkyl, heteroalkyl, heteroaryl, heterocycloalkyl, hydroxy, hydrazino, imino, oxo, nitro, alkylsulfinyl, sulfonic acid, alkylsulfonyl, thiocyanate, thiol, thione, or combinations thereof. By way of example, there may be one, two, three, four, five, or six substitutents. Polymers or similar indefinite structures arrived at by defining substituents with further substituents appended ad infinitum (e.g., a substituted aryl having a substituted alkyl which is itself substituted with a substituted aryl group, which is further substituted by a substituted heteroalkyl group, etc.) are not intended for inclusion herein. Unless otherwise noted, the maximum number of serial substitutions in compounds described herein is three. For example, serial substitutions of substituted aryl groups with two other substituted aryl groups are limited to substituted aryl (substituted aryl) substituted aryl. Similarly, the above definitions are not intended to include impermissible substitution patterns (e.g., methyl substituted with 5 fluorines or heteroaryl groups having two adjacent oxygen ring atoms). Such impermissible substitution patterns are well known to the skilled artisan. When used to modify a chemical group, the term “substituted” may describe other chemical groups defined herein. For example, the term “substituted aryl” includes, but is not limited to, “alkylaryl.” Unless specified otherwise, where is a group is described as optionally substituted, any substituents of the group are themselves unsubstituted.

In some embodiments, the term “substituted alkyl” refers to an alkyl group having one or more substituents including hydroxyl, halo, alkoxy, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl. In additional embodiments, “substituted cycloalkyl” refers to a cycloalkyl group having one or more substituents including alkyl, haloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, halo, hydroxyl; “substituted aryl” refers to an aryl group having one or more substituents including halo, alkyl, haloalkyl, heterocycloalkyl, heteroaryl, alkoxy, and cyano; and “substituted sulfonyl” refers to a group —S(O)2R, in which R is substituted with one or more substituents including alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl. In other embodiments, the one or more substituents may be further substituted with halo, alkyl, haloalkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is substituted. In other embodiments, the substituents may be further substituted with halo, alkyl, haloalkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is unsubstituted.

PI3K Inhibitor Compounds

The present application provides the compounds that function as inhibitors of PI3K isoforms. In one aspect, the PI3K inhibitors are the compounds having the structure of formula (J):

wherein:

n is 0, 1, 2, 3, or 4;

W is CH or N;

A′ is NR5R4, OR4, or NR5C(O)R4;

B′ is alkyl, cycloalkyl, or heteroalkyl, wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl or alkoxy, wherein the heteroalkyl is optionally substituted with optionally substituted alkyl;

R1 is independently selected from halo, cyano, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkoxy, optionally substituted sulfonyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;

R2 is cyano, C3-8 heterocycloalkyl, —NR2xR2x, —NR2xC(O)R2y, —C(O)NR2xR2y, —OR2y, or —SO2R2z; wherein R2x is hydrogen, optionally substituted alkyl, heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl; wherein R2y is hydrogen and alkyl, wherein R2z is alkyl;

R3 is hydrogen, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted cycloalkyl, or optionally substituted aryl;

R4 is heteroaryl optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted alkyl, and —NH2; and

R5 is hydrogen or optionally substituted alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In another aspect, the PI3K inhibitors are the compounds having the structure of formula (I)

n is 0, 1, 2, 3, or 4;

m is 0 or 1;

A is a single bond or C(O);

B is C1-6 alkyl or C3-8 cycloalkyl, wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl or C1-6 alkoxy;

R′ is hydrogen or optionally substituted C1-6 alkyl;

each R1 is independently selected from halo, cyano, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted sulfonyl, optionally substituted C3-8 aryl, optionally substituted C3-8 heteroaryl, optionally substituted C3-8 cycloalkyl, and optionally substituted C2-8 heterocycloalkyl;

R2 is cyano, morpholinyl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z,

    • where R2X is hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, C4-8 heteroaryl;
    • wherein R2y is independently selected from hydrogen and C1-6 alkyl; and
    • wherein R2z is C1-6 alkyl;

R3 is hydrogen, optionally substituted C1-6 alkyl, C3-8 cycloalkyl, or C6-10 aryl;

R4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two heteroatoms selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted C1-6 alkyl, and —NH2; and

R5 is hydrogen or optionally substituted C1-6 alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a four- or eight-membered heterocyclic ring;

    • or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In additional aspect, the compounds have the structure of formula (I) wherein:

n is 0, 1, 2 or 3;

m is 0 or 1;

A is a single bond or C(O);

B is alkyl, cycloalkyl, or heteroalkyl, wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl or alkoxy;

R′ is hydrogen or C1-6 alkyl optionally substituted with C6-10 aryl;

each R1 is independently selected from halo, cyano, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 alkylsulfonyl;

R2 is cyano, morpholinyl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z; wherein R2x is hydrogen, optionally substituted alkyl, heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl; wherein R2y is hydrogen and alkyl, wherein R2z is alkyl;

    • where R2x is hydrogen, C1-6 haloalkyl, C6-10 aryl, C3-8 cycloalkyl, C2-8 heterocycloalkyl, C4-8 heteroaryl, C1-6 alkyl, each of R2x is optionally substituted with one, two, or three members independently selected from halo, C1-6 alkyl, C6-10 aryl, and C3-8 cycloalkyl;
    • wherein R2y is independently selected from hydrogen and C1-6 alkyl; and
    • wherein R2z is C1-6 alkyl;

R3 is hydrogen, C3-8 cycloalkyl, C6-10 aryl, or C1-6 alkyl optionally substituted with C1-6 alkyl or C6-10 aryl, each of which is optionally substituted with C1-6 alkoxy, C1-6 alkyl, or C6-10 aryl;

R4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two heteroatoms selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, C1-6 haloalkyl, C1-6 alkyl, and —NH2; and

R5 is hydrogen or C1-6 alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a four- or eight-membered heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In some other aspect, the compounds have the structure of formula (I), wherein

n is 1 or 2;

m is 0 or 1;

A is a single bond or C(O);

B is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl, each of which is optionally substituted with methoxy, ethoxy, or hydroxyl;

R′ is methyl, ethyl, propyl, phenylmethyl, phenylethyl, or phenylpropyl;

each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl;

R2 is cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, or —SO2R2z;

    • wherein each R2x is independently methyl, butyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl, or oxetanyl, each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, or iodo;
    • wherein each R2y is independently hydrogen, methyl, ethyl, propyl, butyl; and
    • wherein each R2z is independently methyl, ethyl, propyl, or butyl;

R3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, or phenyl;

R4 is a six- to ten-membered heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from fluoro, chloro, bromo, iodo, cyano, —NH2, methyl, ethyl, and propyl; and

R5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (II):

wherein n, A, B′, R1, R2, R3, R4, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (IIa):

wherein n, B′, R1, R2, R3, R4, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (IIb):

wherein n, B′, R1, R2, R3, R4, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (IIc):

wherein n, B′, R1, R2, R3, and R5 are described herein,

p is 0, 1, 2, or 3;

each R4a is independently selected from halo, cyano, C1-4 haloalkyl, and C1-4 alkyl;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (IId):

wherein n, p, B′, R1, R2, R3, R4a, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (III):

wherein n, A, R′, R1, R2, R3, R4, and R5 are described herein,

B is alkyl or cycloalkyl, wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl, alkoxy, or heterocycloalkyl;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (IIIa):

wherein n, B, R′, R1, R2, R3, R4, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formulae (J) or (I) may have the structure of formula (IIIb):

wherein n, p, B, R′, R1, R2, R3, R4a, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In one aspect, the PI3K inhibitors are the compounds having the structure of formula (IV):

wherein:

n is 0, 1, 2, 3, or 4;

W is CH or N;

A′ is NR5R4, OR4, or NR5C(O)R4;

B′ is alkyl, cycloalkyl, heteroalkyl, or heterocycloalkyl wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl or alkoxy, wherein the heteroalkyl is optionally substituted with optionally substituted alkyl;

R1 is independently selected from halo, cyano, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkoxy, optionally substituted sulfonyl, optionally substituted alkylsulfonyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;

R2 is hydrogen, cyano, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, —SO2R2z, optionally substituted C3-8 heterocycloalkyl, or optionally substituted C3-8 heteroaryl; wherein R2x is hydrogen, optionally substituted alkyl, heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl; wherein R2y is hydrogen and alkyl, wherein R2z is alkyl;

R3 is hydrogen, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted cycloalkyl, or optionally substituted aryl;

R4 is heteroaryl optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted alkyl, and —NH2; and

R5 is hydrogen or optionally substituted alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a heterocyclic ring; or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In certain other embodiments, the compounds have the structure of formula (IV)

wherein:

R2 is hydrogen, cyano, morpholinyl, piperidinyl, pyrrolidinyl, piperazinyl optionally substituted with methyl, pyrazolyl, triazolyl, NR2xR2x, NR2yC(O)R2x, C(O)NR2xR2y, OR2y, or SO2R2z wherein

    • R2x is hydrogen, pyrimidinyl, pyridinyl, methyl, ethyl, propyl, tert-butyl, methyl optionally substituted with phenyl or three fluoro, cyclobutyl, cyclohexyl optionally substituted with two fluoro, or oxetanyl;
    • R2y is hydrogen, methyl, tert-butyl;
    • R2z is methyl.

In some other aspect, the compounds have the structure of formula (IV), wherein

n is 1 or 2;

m is 0 or 1;

A is a single bond or C(O);

B is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl, each of which is optionally substituted with methoxy, ethoxy, or hydroxyl;

R′ is methyl, ethyl, propyl, phenylmethyl, phenylethyl, or phenylpropyl;

each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl;

R2 is cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, or —SO2R2z;

    • wherein each R2x is independently hydrogen, methyl optionally substituted with phenyl or three fluoro, ethyl, propyl, butyl, tert-butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl optionally substituted with two fluoro or oxetanyl, or oxetanyl, wherein each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, or iodo;
    • wherein each R2y is independently hydrogen, methyl, ethyl, propyl, butyl, or tert-butyl; and
    • wherein each R2z is independently methyl, ethyl, propyl, or butyl;

R3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, or phenyl;

R4 is a six- to ten-membered heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from fluoro, chloro, bromo, iodo, cyano, —NH2, methyl, ethyl, and propyl; and

R5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In another aspect, the PI3K inhibitors are the compounds having the structure of formula (IVa)

wherein:

n is 0, 1, 2, 3, or 4;

m is 0 or 1;

A is a single bond or C(O);

B is C1-6 alkyl, C3-8 cycloalkyl, C3-8 heterocycloalkyl or C3-8 heterocycloalkyl wherein each of the alkyl, cycloalkyl, heterocycloalkyl, and heterocycloalkyl moieties is optionally substituted with hydroxyl or C1-6 alkoxy;

R′ is hydrogen or optionally substituted C1-6 alkyl;

each R1 is independently selected from halo, cyano, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted sulfonyl, optionally substituted alkylsulfonyl, optionally substituted C3-8 aryl, optionally substituted C3-8 heteroaryl, optionally substituted C3-8 cycloalkyl, and optionally substituted C2-8 heterocycloalkyl;

R2 is hydrogen, cyano, optionally substituted C3-8 heterocycloalkyl, optionally substituted C3-8 heteroaryl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z,

    • where R2x is hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, C4-8 heteroaryl;
    • wherein R2y is independently selected from hydrogen and C1-6 alkyl; and
    • wherein R2z is C1-6 alkyl;

R3 is hydrogen, optionally substituted C1-6 alkyl, C3-8 cycloalkyl, or C6-10 aryl;

R4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two heteroatoms selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted C1-6 alkyl, and —NH2; and

R5 is hydrogen or optionally substituted C1-6 alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a four- or eight-membered heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In certain other embodiments, the compounds have the structure of formula (IVa)

wherein:

n is 0, 1, 2 or 3;

m is 0 or 1;

A is a single bond or C(O);

B is C1-6 alkyl, C3-8 cycloalkyl, C3-8 heteroalkyl or C3-8 heterocycloalkyl wherein each of the alkyl, cycloalkyl, heteroalkyl, and heterocycloalkyl moieties is optionally substituted with hydroxyl or C1-6 alkoxy;

R′ is hydrogen or C1-6 alkyl optionally substituted with C6-10 aryl;

each R1 is independently selected from halo, cyano, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 alkylsulfonyl;

R2 is hydrogen, cyano, morpholinyl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, phenyl, pyrazolyl, or triazolyl.

    • where R2x is hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, C4-8 heteroaryl;
    • wherein R2y is independently selected from hydrogen and C1-6 alkyl; and
    • wherein R2z is C1-6 alkyl;

R3 is hydrogen, optionally substituted C1-6 alkyl, C3-8 cycloalkyl, or C6-10 aryl;

R4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two heteroatoms selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted C1-6 alkyl, and —NH2; and

R5 is hydrogen or optionally substituted C1-6 alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a four- or eight-membered heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In certain other embodiments, the compounds have the structure of formula (IVa)

wherein:

n is 1 or 2;

m is 0 or 1;

A is a single bond or C(O);

B is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with methoxy, ethoxy, or hydroxyl;

R′ is methyl, ethyl, propyl, phenylmethyl, phenylethyl, or phenylpropyl;

each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl;

R2 is hydrogen, cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, —SO2R2z, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, phenyl, pyrazolyl, or triazolyl.

    • wherein each R2x is independently hydrogen, methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl, or oxetanyl, each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, or iodo;
    • wherein each R2y is independently hydrogen, methyl, ethyl, propyl, butyl; and
    • wherein each R2z is independently methyl, ethyl, propyl, or butyl;

R3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, or phenyl;

R4 is a six- to ten-membered heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from fluoro, chloro, bromo, iodo, cyano, —NH2, methyl, ethyl, and propyl; and

R5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In one embodiment, the compounds of formula (IV) may have the structure of formula (IVb):

wherein n, A, R′, R1, R2, R3, R4, and R5 are described herein,

B is alkyl or cycloalkyl, wherein each of the alkyl and cycloalkyl moieties is optionally substituted with hydroxyl, alkoxy, or heterocycloalkyl;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In some other embodiments, the compounds have the structure of formula (IVb)

wherein:

n is 1 or 2;

A is a single bond or C(O);

B is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with methoxy, ethoxy, or hydroxyl;

R′ is methyl, ethyl, propyl, phenylmethyl, phenylethyl, or phenylpropyl;

each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl;

R2 is hydrogen, cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, —SO2R2z, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, phenyl, pyrazolyl, or triazolyl.

    • wherein each R2x is independently hydrogen, methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl, or oxetanyl, each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, or iodo;
    • wherein each R2y is independently hydrogen, methyl, ethyl, propyl, butyl; and
    • wherein each R2z is independently methyl, ethyl, propyl, or butyl;

R3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, or phenyl;

R4 is a six- to ten-membered heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from fluoro, chloro, bromo, iodo, cyano, —NH2, methyl, ethyl, and propyl; and

R5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formula (IV) may have the structure of formula (IVc):

wherein n, B, R′, R1, R2, R3, R4, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In some other embodiments, the compounds have the structure of formula (IVc)

wherein:

n is 1 or 2;

B is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with methoxy, ethoxy, or hydroxyl;

R′ is methyl, ethyl, propyl, phenylmethyl, phenylethyl, or phenylpropyl;

each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl;

R2 is hydrogen, cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, —SO2R2z, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, phenyl, pyrazolyl, or triazolyl.

    • wherein each R2x is independently hydrogen, methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl, or oxetanyl, each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, or iodo;
    • wherein each R2y is independently hydrogen, methyl, ethyl, propyl, butyl; and
    • wherein each R2z is independently methyl, ethyl, propyl, or butyl;

R3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, or phenyl;

R4 is a six- to ten-membered heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from fluoro, chloro, bromo, iodo, cyano, —NH2, methyl, ethyl, and propyl; and

R5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

The compounds of formula (IV) may have the structure of formula (IVd):

wherein n, p, B, R′, R1, R2, R3, R4a, and R5 are described herein,

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In some other embodiments, the compounds have the structure of formula (IVd)

wherein:

n is 1 or 2;

R′ is methyl, ethyl, propyl, phenylmethyl, phenylethyl, or phenylpropyl;

each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl;

R2 is hydrogen, cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, —SO2R2z, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, phenyl, pyrazolyl, or triazolyl.

    • wherein each R2x is independently hydrogen, methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl, or oxetanyl, each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, or iodo;
    • wherein each R2y is independently hydrogen, methyl, ethyl, propyl, butyl; and
    • wherein each R2z is independently methyl, ethyl, propyl, or butyl;

R3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, or phenyl;

R4a is cyano, —NH2, fluoro, chloro, bromo, iodo, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, or trifluoroethyl;

R5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

In one embodiment, the PI3K inhibitors are compounds having the structures of any of the foregoing formulae (IIc), (IId), and (IIIb), wherein n, R1, R2, R3, and R5 are described herein; p is 2 or 3; and each R4a is independently selected from cyano, —NH2, fluoro, chloro, bromo, iodo, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, or trifluoroethyl. In some embodiment, the PI3K inhibitors are compounds having the structures of formula (IVd), wherein n, R1, R2, R3, and R5 are described herein; p is 2 or 3; and each R4a is independently selected from cyano, —NH2, fluoro, chloro, bromo, iodo, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, or trifluoroethyl. In other embodiment, the PI3K inhibitors are compounds having the structures of any of the foregoing formulae (J), (II), (IIa), (IIb), (IIc), and (IId), wherein B′ is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members independently selected from cyano, —OR2y, or —SO2R2z, wherein R2y is hydrogen, methyl, ethyl, propyl, butyl, or pentyl, and wherein R2z is methyl, ethyl, propyl, butyl, or pentyl. In other embodiment, the PI3K inhibitors are compounds having the structures of formula (IV), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members independently selected from cyano, —OR2y or —SO2R2z, wherein R2y is hydrogen, methyl, ethyl, propyl, butyl, or pentyl, and wherein R2z is methyl, ethyl, propyl, butyl, or pentyl. In some embodiments, the PI3K inhibitors are compounds having the structures of any of the foregoing formulae (J), (II) (IIa), (IIb), (IIc), and (IId), wherein B′ is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one member of —SO2R2z, wherein R2z is methyl, ethyl, propyl, butyl, or pentyl. In certain embodiments, the PI3K inhibitors are compounds having the structures of any of the foregoing formulae (J), (II) (IIa), (IIb), (IIc), and (IId), wherein B′ is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members of —OR2y, wherein R2z is hydrogen, methyl, ethyl, propyl, butyl, or pentyl. In other embodiments, compounds having the structures of any of the foregoing formulae (J), (II) (IIa), (IIb), (IIc), and (IId) wherein B′ is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one member of cyano. In other embodiment, the PI3K inhibitors are compounds having the structures of formula (IV), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members independently selected from cyano, —OR2y, or —SO2R2z, wherein R2y is hydrogen, methyl, ethyl, propyl, butyl, or pentyl, and wherein R2z is methyl, ethyl, propyl, butyl, or pentyl. In certain embodiments, the PI3K inhibitors are compounds having the structures of formula (IV), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members of —OR2y, wherein R2z is hydrogen, methyl, ethyl, propyl, butyl, or pentyl. In other embodiments, compounds having the structures of formulae (IV) wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one member of cyano. In other embodiment, the PI3K inhibitors are compounds having the structures of formulae (I), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members independently selected from cyano, —OR2y, or —SO2R2z, wherein R2y is hydrogen, methyl, ethyl, propyl, butyl, or pentyl, and wherein R2z is methyl, ethyl, propyl, butyl, or pentyl. In some embodiments, the PI3K inhibitors are compounds having the structures of formulae (I) wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one member of —SO2R2z, wherein R2z is methyl, ethyl, propyl, butyl, or pentyl. In certain embodiments, the PI3K inhibitors are compounds having the structures of formulae (I), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members of —OR2y, wherein R2z is hydrogen, methyl, ethyl, propyl, butyl, or pentyl. In other embodiments, compounds having the structures of any of the foregoing formulae (I), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one member of cyano. In other embodiment, the PI3K inhibitors are compounds having the structures of any of the foregoing formulae (IVa), (IVb), (IVc), and (IVd), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members independently selected from cyano, —OR2y, or —SO2R2z, wherein R2y is hydrogen, methyl, ethyl, propyl, butyl, or pentyl, and wherein R2z is methyl, ethyl, propyl, butyl, or pentyl. In certain embodiments, the PI3K inhibitors are compounds having the structures of the foregoing formulae (IVa), (IVb), (IVc), and (IVd), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one or two members of —OR2y, wherein R2z is hydrogen, methyl, ethyl, propyl, butyl, or pentyl. In other embodiments, compounds having the structures of any of the foregoing formulae (IVa), (IVb), (IVc), and (IVd), wherein B is C1-6 alkyl or C3-8 cycloalkyl, each of which is substituted with one member of cyano.

In one embodiment, n is 0. In some embodiments, n is 1, 2, 3, or 4. In some embodiments, n is 1, 2, or 3. In other embodiments, n is 1 or 2. In certain embodiments, n is 1 and the R1 moiety may be located on any position of the phenyl of the quinazolinone ring. In another embodiment, n is 2. Both R1 substituents or moieties may be the same or different. Two R1 moieties may be located on any two positions of the phenyl of the quinazolinone ring. By way of example, the first R1 may be ortho, meta, or para to the second R1. In yet another embodiment, n is 3. All the R1 substituents or moieties may be the same or different, or two R1 may be the same and different from the third R1. Three R1 moieties may be located on any three positions of the phenyl of the quinazolinone ring. For example, the first R1 may be ortho to the second R1, and the first R1 may be para to the third R1. In yet another embodiment, n is 4. All the R1 substituents may be the same or different, three R1 may be the same and different from the fourth R1, two R1 may be the same and different from the third and the fourth R1.

In some other embodiments, each R1 is independently halo, cyano, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, hydroxy, optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substituted C6-10 aryl, optionally substituted C4-8 heteroaryl, or optionally substituted C1-6 alkylsulfonyl. In certain embodiments, each R1 is independently halo, cyano, optionally substituted C1-4 alkyl, optionally substituted C1-4 haloalkyl, optionally substituted C1-4 alkoxy, optionally substituted C3-6 cycloalkyl, or optionally substituted C1-4 alkylsulfonyl. In other embodiments, each R1 is independently halo, cyano, C1-4 haloalkyl, C1-4 alkyl, or C1-4 alkylsulfonyl. In certain embodiments, each R1 is independently selected from fluoro, chloro, iodo, bromo, cyano, methyl, ethyl, propyl, butyl, fluoromethyl, fluoroethyl, difluoromethyl, difluoroethyl, trifluoromethyl, trifluoroethyl, methylsulfonyl, ethylsulfonyl, or propylsulfonyl. In some embodiments, each R1 is independently fluoro, chloro, bromo, iodo, cyano, methyl, ethyl, difluoromethyl (—CHF2), trifluoromethyl (—CF3), methoxy, methylsulfonyl (—SO2CH3), cyclopropylmethyl, or cyclopropyl. In certain embodiments, each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, or propyl. In one embodiment, each R1 is independently fluoro, chloro, cyano, methylsulfonyl, methyl, or trifluoromethyl.

In one embodiment, m is 0. In some embodiments, m is 1. In the embodiment where m is 1, the compounds of the present application may have the structures of any of formulae (III), (IIIa), and (IIIb). In the embodiments where m is 1, the N atom of the NR′ moiety is attached to (or connects to) the quinazolinone ring. R′ is hydrogen or C1-6 alkyl optionally substituted with C6-10 aryl. In some embodiments, R′ is hydrogen, methyl, ethyl, propyl, butyl, pentyl, hexyl, phenylmethyl, phenylethyl, or phenylpropyl. R′ is hydrogen methyl, ethyl, propyl, phenylmethyl, or phenylethyl.

The present application provides that R2 is cyano, morpholinyl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z; where R2x is hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, C4-8 heteroaryl; where R2y is independently selected from hydrogen and C1-6 alkyl; wherein R2z is C1-6 alkyl; and each of R2x, R2y, and R2z is optionally substituted with one, two, or three members of halogens. Additionally, R2 is hydrogen, cyano, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, —SO2R2z, optionally substituted C3-8 heterocycloalkyl, or optionally substituted C3-8 heteroaryl; where R2x is hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, C4-8 heteroaryl; where R2y is independently selected from hydrogen and C1-6 alkyl; wherein R2z is C1-6 alkyl; and each of R2x, R2y, and R2z is optionally substituted with one, two, or three members of halogens. In some embodiment, R2 is hydrogen, cyano, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z, morpholinyl, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, phenyl, pyrazolyl, or triazolyl; where R2x is hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, C4-8 heteroaryl; where R2y is independently selected from hydrogen and C1-6 alkyl; wherein R2z is C1-6 alkyl; and each of R2x, R2y, and R2z is optionally substituted with one, two, or three members of halogens. In certain embodiments, R2 is hydrogen, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, phenyl, pyrazolyl, or triazolyl. In certain other embodiments, R2 is hydrogen, pyrrolidinyl, piperidinyl, phenyl, pyrazolyl, or triazolyl piperazinyl optionally substituted with C1-6 alkyl. In additional embodiments, R2 is hydrogen, pyrrolidinyl, piperidinyl, phenyl, pyrazolyl, or triazolyl piperazinyl optionally substituted with methyl, ethyl, propyl, or butyl. In certain embodiments, R2 is cyano, morpholinyl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2x, —OR2y, —SO2R2z, wherein R2x is hydrogen, C1-6 alkyl optionally substituted with C6-10 aryl, C1-6 haloalkyl, C6-10 aryl, C3-8 cycloalkyl optionally substituted with one to three members of halogen atoms, C2-8 heterocycloalkyl, or C4-8 heteroaryl; R2y is independently selected from hydrogen and C1-6 alkyl; and R2z is C1-6 alkyl. In further embodiments, R2 is cyano, —NH2, —NHR2x (wherein R2x is methyl, butyl, propyl, phenylmethyl, phenylethyl, phenyl, pyridinyl, pyrimidinyl, cyclobutyl, oxetanyl, cyclohexyl which is optionally substituted with one or two fluoro atoms), —NHC(O)H, —NHC(O)R2y (wherein R2y is methyl, ethyl, propyl, or butyl), NR2xR2y (where R2x is methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenylmethyl, or phenylethyl; and R2y is methyl, ethyl, or propyl), —OH, —OR2y (wherein R2y is methyl, ethyl, propyl, or butyl), and —SO2NHR2x (wherein R2z is methyl, ethyl, propyl, or butyl). In further other embodiments, R2 is cyano, morpholinyl, —NH2, —NHCH3, —NHCH2CH3, —NHC2H5, —NH(CH2)2CH3, —NHCH(CH3)2, —NH(CH2)3CH3, —NH(CH2)4CH3, —NH(CH2)5CH3, —NHC(CH3)3, —NHCH2C6H5, NH-pyridinyl, NH-pyrimidinyl, —NHcyclopropyl (i.e. —NHC3H5), —NHcycylobutyl (i.e. —NHC4H7), —NHcyclopentyl (i.e. —NHC6H11), —NHcyclohexyl (i.e. —NHC6H11) substituted with two fluoride atoms (which may also be referred to as —NH(difulorocyclohexyl)), —NHoxetanyl (i.e. NHC3H5O), —NHC(O)CH3, —NHC(O)C2H5, —NHC(O)H, —N(CH3)(CH3), —N(CH3)(CH2CF3), —N(CH3)(CH2C6H5), —OH, —OCH3, —OC2H5, —CONH2, CON(CH3)(CH3), —SO2CH3, —SO2C2H5, and —SO2C3H7.

In the present application, B is C1-6 alkyl optionally substituted with hydroxyl, C1-6 alkoxy, or C3-8 cycloalkyl; C3-8 cycloalkyl optionally substituted with hydroxyl or C1-6 alkoxy; or C1-6 heteroalkyl optionally substituted with C1-6 alkyl or C6-10 arylC1-6 alkyl. In some embodiments, B is C1-6 alkyl optionally substituted with one, two, or three members independently selected from hydroxyl and C1-6 alkoxy. In certain embodiments, B′ is C3-8 cycloalkyl optionally substituted with one, two, or three members independently selected from hydroxyl and C1-6 alkoxy. In other embodiments, B′ is C1-6 alkyl or C3-8 cycloalkyl, each of the alkyl or cycloalkyl moieties is optionally substituted with one, two, or three members independently selected from hydroxyl and C1-6 alkoxy. In other embodiments, B′ is C1-6 heteroalkyl optionally substituted with one, two, or three members selected from C1-6 alkyl and C6-10 arylC1-6 alkyl. In some other embodiments, B′ is C1-6 alkyl, which is optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, and hydroxyl. In yet some other embodiments, B′ is C3-8 cycloalkyl optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, and hydroxyl. In additional other embodiments, B′ is C1-6 haloalkyl optionally substituted with one, two, or three members independently selected from methyl, ethyl, propyl, phenylmethyl, phenylethyl, and phenylpropyl. In further embodiments, B′ is methyl, ethyl, propyl, butyl, pentyl, or hexyl, each of which is optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, and hydroxyl. In other further embodiments, B′ is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, each of which is optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, and hydroxyl. In additional other embodiments, B′ is C1-6 haloalkyl having nitrogen atom, wherein carbon atoms of the haloalkyl moiety is optionally substituted with one, two, or three members independently selected from methyl, ethyl, propyl, phenylmethyl, phenylethyl, and phenylpropyl.

Also in the present application, B is C1-6 alkyl or C3-8 cycloalkyl, each of which is optionally substituted with hydroxyl or C1-6 alkoxy. In some embodiments, B is C1-6 alkyl optionally substituted with one, two, or three members independently selected from hydroxyl and C1-6 alkoxy. In certain embodiments, B is C3-8 cycloalkyl optionally substituted with one, two, or three members independently selected from hydroxyl and C1-6 alkoxy. In some other embodiments, B is C1-6 alkyl, which is optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, or hydroxyl. In yet some other embodiments, B is C3-8 cycloalkyl optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, and hydroxyl. In further embodiments, B is methyl, ethyl, propyl, butyl, pentyl, or hexyl, each of which is optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, and hydroxyl. In other further embodiments, B is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, each of which is optionally substituted with one, two, or three members independently selected from methoxy, ethoxy, and hydroxyl. Each and every variation of B or B′ may be combined with each and every variation of n, m, R′, R1, R2, R3, R4, and R5 as described herein. It is understood that the divalent groups B and B′ may be represented by the alkyl terms as defined above. For example, B is methylene, ethylene, propylene, butylene, pentylene, hexylene, cyclopropylene, cyclobutylene, cyclopentylene, or cyclohexylene may be represented by methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, respectively.

In certain embodiments, R3 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C3-8 cycloalkyl, or optionally substituted C6-10 aryl. In one embodiment, R3 is hydrogen, C3-6 cycloalkyl, C6-10 aryl, or C1-6 alkyl optionally substituted with hydroxy, C6-10 arylC1-6 alkoxy, or C3-6 cycloalkyl. In some embodiments, R3 is hydrogen, methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropylmethyl, cyclopropylethyl, cyclobutylmethyl, cyclobutylethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CH2OH, —C2H4OH, —C3H6OH, benzyloxymethyl (i.e,


or phenyl (i.e.


In some other embodiments, R3 is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, or phenyl.

In additional embodiments, R5 is hydrogen or optionally substituted C1-6 alkyl. In some embodiments, R5 is hydrogen or C1-4 alkyl. In certain embodiments, R5 is hydrogen, methyl, ethyl, propyl or butyl. In certain other embodiments, R5 is hydrogen.

In further embodiments, R3 and R5 with the atoms to which they are attached (e.g. carbon and nitrogen, respectively) optionally form a heterocyclic ring. In some embodiments, the R3-R5 heterocyclic ring is optionally substituted with fluoro, chloro, bromo, or iodo. In other embodiments, the R3-R5 heterocyclic ring is a three- to eight-membered heterocycloalkyl (i.e. heterocycloalkyl having three to eight ring members and at least one ring member is a heteroatom). In other embodiments, the R3-R5 heterocyclic ring is a four- to seven-membered heterocycloalkyl (i.e. heterocycloalkyl having four to seven ring members and at least one ring member is a heteroatom). In one embodiment, the R3-R5 heterocyclic ring is a five-membered heterocycloalkyl. In certain other embodiments, the R3-R5 heterocyclic ring is C3-8 heterocycloalkyl. In certain embodiments, the R3-R5 heterocyclic ring is azepanyl, azetidinyl, piperidinyl, or pyrrolidinyl. In some other embodiments, the R3-R5 heterocyclic ring is pyrrolidinyl. In one other embodiment, the R3-R5 heterocyclic ring is a five-membered heterocycloalkyl substituted with halo. In other additional embodiments, the R3-R5 heterocyclic ring is pyrrolidinyl substituted with fluoro, chloro, bromo, or iodo.

In one embodiment, R4 is heteroaryl having at least two nitrogen atoms and at least one aromatic ring, and R4 heteroaryl is optionally substituted one, two, or three members independently selected from halo, cyano, —NH2, C1-6 alkyl, and C1-6 haloalkyl. In certain embodiments, R4 heteroaryl is a six- to twelve-membered heteroaryl (i.e. heteroaryl having six to twelve ring members). In certain other embodiments, R4 heteroaryl is a six- to ten-membered heteroaryl (i.e. heteroaryl having six to ten ring members). R4 heteroaryl may be a monocyclic or bicyclic heteroaryl. In some embodiments, R4 heteroaryl is a monocyclic heteroaryl having at least two nitrogen atoms. In certain embodiments, R4 heteroaryl is a bicyclic heteroaryl having at least one aromatic ring, at least two nitrogen atoms, and at least one additional heteroatom selected from N, O, or S. In certain other embodiments, R4 is selected from purinyl, pyrimidinyl, thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrimidinyl, or imidazotriazinyl.

In any of the foregoing formulae, R4 is heteroaryl optionally substituted with one, two, or three members independently selected from halo, cyano, —NH2, C1-6 alkyl, and C1-6 haloalkyl, wherein the R4 heteroaryl is selected from the group consisting of

In some other embodiments, R4 is optionally substituted with one to three members independently selected from fluoro, chloro, bromo, iodo, cyano, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, and —NH2. In yet other embodiments, R4 is optionally substituted with one to three members independently selected from fluoro, chloro, and bromo.

In certain other embodiments, R4 is selected from purinyl, pyrimidinyl, thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrimidinyl, and imidazotriazinyl, each of which is optionally substituted with one, two, or three members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and —NH2. In certain other embodiments, R4 is selected from thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrimidinyl, and imidazotriazinyl, each of which is optionally substituted with one or two members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and —NH2. In other embodiments, R4 is selected from thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrimidinyl, and imidazotriazinyl, each of which is optionally substituted with independently chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and —NH2. In other embodiments, R4 is selected from thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrimidinyl, and imidazotriazinyl, each of which is optionally substituted with independently with one member selected from chloro, fluoro, bromo, and iodo. In other embodiments, R4 is selected from thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrimidinyl, and imidazotriazinyl, each of which is optionally substituted with one or two members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and —NH2. In certain other embodiments, R4 is pyrimidinyl or pyrazinyl, each substituted with two or three members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and —NH2. Each and every variation of n, m, R′, R2, R1, R2, R3, R4, and R5 may be combined with each and every variation thereof as described above.

In some embodiments, A is C(O). In certain embodiments, A is a single bond. Each and every variation of A may be combined with each and every variation of n, m, R′, R1, R2, R3, R4, and R5 as described above. In some other embodiments, A is C(O) and R4 heteroaryl is a bicyclic group defined above. In other embodiments, A is a single bond and R4 heteroaryl is a bicyclic group defined above. In additional embodiments, A is C(O) and R4 heteroaryl is a monocyclic group selected from pyrimidinyl and pyrazinyl. In other embodiments, A is a single bond and R4 heteroaryl is a monocyclic group selected from pyrimidinyl and pyrazinyl.

In further embodiments, A′ is OR4. In some further embodiments, A′ is N(R5)C(O)R4. In yet further embodiments, A′ is NR5R4. Each and every variation of A′ may be combined with each and every variation of n, m, R2′, R1, R3, R4, and R5 as described above.

In certain embodiments, W is CH or N. In certain other embodiments, W is CH. In yet other embodiments, W is N. Each and every variation of W may be combined with each and every variation of n, m, A′, R2′, R1, R2, R3, R4, and R5 as described above.

In the formulae (IIc), (IId), (IIIa), and (IIIb), n, R1, R2, R3, and R5 as described herein; p is 0, 1, 2, or 3; and R4a is independently selected from halo, cyano, —NH2, and optionally substituted C1-6 alkyl. In the formula (IVd), n, R1, R2, R3, and R5 as described herein; p is 0, 1, 2, or 3; and R4a is independently selected from halo, cyano, —NH2, and optionally substituted C1-6 alkyl. In one embodiment, p is 0. In certain embodiments, p is 1, 2, or 3. In some embodiments, p is 1 or 2. In some embodiments, p is 1 and R4a moiety may be located on any position of the pyrimdinyl or pyrazinyl ring. In another embodiment, p is 2 and both R4a substituents or moieties may be the same or different; each of the two R4a moiety may be located on any position of the pyrimdinyl or pyrazinyl ring. In yet another embodiment, p is 3 and all R4a substituents may be the same or different, or two R4a may be the same and different from the third R4a.

In the present application, each R4a is independently selected from halo, —NH2, cyano, C1-6 haloalkyl, and C1-6 alkyl. In some embodiments, each R4a is independently selected from fluoro, chloro, bromo, iodo, cyano, —NH2, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, and difluoroethyl. In additional embodiments, p is 2 or 3, and each R4a is independently selected from fluoro, chloro, bromo, iodo, cyano, —NH2, methyl, and ethyl. Each and every variation of p and R4a may be combined with each and every variation of n, R′, R1, R2, R3, and R5 as described above.

The compounds of the present application may bear one or more chiral centers. The compounds bearing the chiral center have the same molecular formula and the same chemical name with different stereoisomer designations. For example, the below 2,4-diamino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile bearing one chiral center can be resolved into the (S) and (R) enantiomers, (S)-2,4-diamino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile and (R)-2,4-diamino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile.

Representative compounds of the present application are listed in Table 1 below. Additional compounds are listed in Table 1a. The compounds may be named using the nomenclature systems and symbols that are commonly recognized in the art of chemistry including, for example, ChemBioDraw Ultra 12.0, Chemical Abstract Service (CAS), and International Union of Pure and Applied Chemistry (IUPAC). For example, compound 42 in table 1 may be named as tert-butyl N-[3-[5-chloro-2-[(1S)-1-[(2,6-diamino-5-cyano-pyrimidin-4-yl)amino]ethyl]-4-oxo-quinazolin-3-yl]propyl]carbamateor (S)-2,4-diamino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile by using IUPAC or ChemBioDraw Ultra 12.0, respectively.

TABLE 1 Representative Compounds compound structure 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 52 51 53 54 55 56 57 58 59 60 61 62 63 64 65 66 68 69 71 72 73 74 75

TABLE 1a Representative Compounds compound structure 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 140 142 143

The present application provides pharmaceutically acceptable salts, hydrates, solvates, isomers, tautomers, stereoisomers, enantiomers, racemates, atropisomers, polymorphs, prodrugs, or a mixture thereof, of the compounds described herein. In addition, the present application provides the compounds in which from 1 to n hydrogen atoms attached to a carbon atom may be replaced by a deuterium atom or D, in which n is the number of hydrogen atoms in the molecule. It is known that the deuterium atom is a non-radioactive isotope of the hydrogen atom. Such compounds may increase resistance to metabolism, and thus may be useful for increasing the half-life of the compounds of any of the formulae described herein or pharmaceutically acceptable salts, isomers, prodrugs, or solvates thereof, when administered to a mammal. See, e.g., Foster, “Deuterium Isotope Effects in Studies of Drug Metabolism”, Trends Pharmacol. Sci., 5(12):524-527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogen atoms have been replaced by deuterium.

The terms “a compound of the present application,” “a compound described herein,” “a compound of any of the formulae described herein,” or variant thereof refer to a compound having the structure of any of the formulae (J), (I), (II), (IIa), (IIb), (IIc), (IId), (III), (IIIa), and (IIIb). The terms “a compound of the present application,” “a compound described herein,” “a compound of any of the formulae described herein,” or variant thereof also refer to a compound having the structure of any of the formulae (IV), (IVa), (IVb), (IVc), and (IVd). In one embodiment, compounds of the present application are Compounds 1-143 as described herein.

“Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use. “Pharmaceutically acceptable salts” or “physiologically acceptable salts” refer to salts of pharmaceutical compounds that retain the biological effectiveness and properties of the underlying compound, and which are not biologically or otherwise undesirable. There are acid addition salts and base addition salts. Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Acids and bases useful for reaction with an underlying compound to form pharmaceutically acceptable salts (acid addition or base addition salts respectively) are known to one of skill in the art. Similarly, methods of preparing pharmaceutically acceptable salts from an underlying compound (upon disclosure) are known to one of skill in the art and are disclosed in for example, Berge, at al. Journal of Pharmaceutical Science, January 1977 vol. 66, No. 1, and other sources. If the compounds described herein are obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt.

Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.

“Isomers” refers to compounds that have the same molecular formula. As used herein, the term isomers include double bond isomers, racemates, stereoisomers, enantiomers, diastereomers, and atropisomers. Single isomers, such as enantiomers or diastereomers, can be obtained by asymmetric synthesis or by resolution of a mixture of isomers. Resolution of a mixture of isomers (e.g. racemates) maybe accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral high pressure liquid chromatography (HPLC) column. “Double bond isomers” refer to Z- and E-forms (or cis- and trans-forms) of the compounds with carbon-carbon double bonds.

“Atropisomers” refers to conformational stereoisomers which occur when rotation about a single bond in the molecule is prevented, or greatly hindered, as a result of steric interactions with other parts of the molecule and the substituents at both ends of the single bond are asymmetrical, i.e., they do not require a stereocenter. Where the rotational barrier about the single bond is high enough, and interconversion between conformations is slow enough, separation and isolation of the isomeric species may be permitted. Atropisomers may be separated by the methods well known in the art. Unless otherwise indicated, the description is intended to include individual atropisomers as well as mixtures. Also, as understood by those skilled in the art, the atropisomers may be represented by the same chemical name with different atropisomer designations. By way of example, the below structures are atropisomers of compound 11, (S)-3-((2-aminoethyl)(methyl)amino)-5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)quinazolin-4(3H)-one.

“Racemates” refers to a mixture of enantiomers.

“Stereoisomers” or “stereoisomeric forms” refer to compounds that differ in the chirality of one or more stereocenters. Stereoisomers include enantiomers and diastereomers. The compounds may exist in stereoisomeric form if they possess one or more asymmetric centers or a double bond with asymmetric substitution and, therefore, can be produced as individual stereoisomers or as mixtures. Unless otherwise indicated, the description is intended to include individual stereoisomers as well as mixtures. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art (see, e.g., Chapter 4 of Advanced Organic Chemistry, 4th ed., J. March, John Wiley and Sons, New York, 1992).

“Tautomers” or “tautomeric formers” refer to alternate forms of a compound that differ in the position of a proton, such as enol-keto and imine-enamine tautomers, or heteroaryls such as pyrazoles, imidazoles, benzimidazoles, triazoles, and tetrazoles.

A “solvate” is formed by the interaction of a solvent and a compound. Solvates of salts of the compounds of any of the formulae described herein are also provided. Hydrates of the compounds of any of the formulae are also provided.

A “prodrug” is defined in the pharmaceutical field as a biologically inactive derivative of a drug that upon administration to the human body is converted to the biologically active parent drug according to some chemical or enzymatic pathway.

In any one of the foregoing embodiments, the compound described herein or a pharmaceutically acceptable salt thereof is an (S)-enantiomer. In any one of the foregoing embodiments, the compound described herein or a pharmaceutically acceptable salt thereof is an (R)-enantiomer. In any one of the foregoing embodiments, the compound described herein or a pharmaceutically acceptable salt thereof is an atropisomer.

The application also provides a composition containing a mixture of enantiomers of the compound or a pharmaceutically acceptable salt thereof. In one embodiment, the mixture is a racemic mixture. In other embodiments, the composition comprises the (S)-enantiomer of a compound in excess of over the corresponding the (R)-enantiomer of the compound. In some embodiments, the composition contains the (S)-enantiomer of the compound and is substantially free of its corresponding (R)-enantiomer. In certain embodiments, a composition substantially free of the (R)-enantiomer has less than or about 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5%, 1%, 0.05%, or 0.01% of the (R)-enantiomer. In other embodiments, the composition containing the (S)-enantiomer of a compound or a pharmaceutically acceptable salt thereof, predominates over its corresponding (R)-enantiomer by a molar ratio of at least or about 9:1, at least or about 19:1, at least or about 40:1, at least or about 80:1, at least or about 160:1, or at least or about 320:1.

The composition containing a compound according to any of the formulae described herein or a pharmaceutically acceptable salt thereof, may also contain the compound in enantiomeric excess (e.e.). By way of example, a compound with 95% (S)-isomer and 5% (R)-isomer will have an e.e. of 90%. In some embodiments, the compound has an e.e. of at least or about 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%.

In any one of the foregoing embodiments, the compound or a pharmaceutically acceptable salt thereof, is an atropisomer. Another embodiment provides the composition containing a mixture of atropisomers of the compound or a pharmaceutically acceptable salt thereof. By way of example, a compound with 95% of one atropisomer and 5% of the other atropisomers. In some embodiments, a compound with about 90, 80, 70, 60, 50, 40, 30, 20, or 10% of one atropisomer and 10, 20, 30, 40, 50, 60, 70, 80, or 90%, respectively, of the other atropisomers.

The application also provides the free base forms of the compounds described herein. In certain embodiments, provided herein are the enantiomers, (R) or (S), of the compounds of the formulae described herein. In other embodiments, provided herein are the atropisomers of the compounds of the formulae described herein.

The application further provides compositions comprising the compounds described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof. The composition may include racemic mixtures, mixtures containing an enantiomeric excess of one enantiomer or single diastereomers or diastereomeric mixtures. All such isomeric forms of these compounds are expressly included herein, the same as if each and every isomeric form were specifically and individually listed.

In certain embodiments, provided herein are also polymorphs, such as crystalline and amorphous forms, of the compounds described herein. In some embodiments, provided are also chelates, non-covalent complexes, and mixtures thereof, of the compounds of the formula described herein or pharmaceutically acceptable salts, prodrugs, or solvates thereof. A “chelate” is formed by the coordination of a compound to a metal ion at two (or more) points. A “non-covalent complex” is formed by the interaction of a compound and another molecule wherein a covalent bond is not formed between the compound and the molecule. For example, complexation can occur through van der Waals interactions, hydrogen bonding, and electrostatic interactions (also called ionic bonding).

Therapeutic Uses of the Compounds

The compounds of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof may be used for the treatment of diseases and/or conditions mediated by PI3K isoforms. In addition, the application provides the compounds for use in therapy. Also, provided herein are methods for inhibiting one or more PI3K isoforms. In one embodiment, provided are methods for inhibiting PI3Kδ activity using the compound described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof. In other embodiment, provided are methods for inhibiting PI3Kδ and/or PI3Kβ activities using the compound or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof. The application further provides methods for use in such methods. The PI3K isoforms may be selectively or specifically inhibited. Additionally, the compounds may be used to inhibit PI3K activity therapeutically or prophylactically, such as PI3Kδ and/or PI3Kβ.

The compounds according to the present application may be used in combination with one or more additional therapeutic agents. The therapeutic agents may be in the forms of compounds, antibodies, polypeptides, or polynucleotides. The therapeutic agent includes, but is not limited to, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof. In one embodiment, the application provides a product comprising a compound described herein and an additional therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy, e.g. a method of treating a disease, disorder, or condition that is mediated by PI3K isoforms.

Also, the therapeutic agents may be those that inhibit or modulate the activities of Bruton's tyrosine kinase, spleen tyrosine kinase, apoptosis signal-regulating kinase, Janus kinase, lysyl oxidase, lysyl oxidase-like proteins, matrix metallopeptidase, bromodomain-containing protein, adenosine A2B receptor, isocitrate dehydrogenase, serine/threonine kinase TPL2, discoidin domain receptor, serine/threonine-protein kinases, IKK, MEK, EGFR, histone deacetylase, protein kinase C, or any combination thereof. In certain embodiments, the therapeutic agent may be selected from a PI3K (including PI3Kγ, PI3Kδ, PI3Kβ, PI3Kα, and/or pan-PI3K) inhibitor, a JAK (Janus kinase, including JAK1, JAK2, and/or JAK3) inhibitor, a SYK (spleen tyrosine kinase) inhibitor, a BTK (Bruton's tyrosine kinase) inhibitor, an A2B (adenosine A2B receptor) inhibitor, an ACK (activated CDC kinase, including ACK1) inhibitor, an ASK (apoptosis signal-regulating kinase, including ASK1) inhibitor, Auroa kinase, a BRD (bromodomain-containing protein, including BRD4) inhibitor, a Bcl (B-cell CLL/lymphoma, including Bcl-1 and/or Bcl-2) inhibitor, a CAK (CDK-activating kinase) inhibitor, a CaMK (calmodulin-dependent protein kinases) inhibitor, a CDK (cyclin-dependent kinases, including CDK1, 2, 3, 4, and/or 6) inhibitor, a CK (casein kinase, including CK1 and/or CK2) inhibitor, a DDR (discoidin domain receptor, including DDR1 and/or DDR2) inhibitor, a EGFR inhibitor, a FXR (farnesoid x receptor) inhibitor, a FAK (focal adhesion kinase) inhibitor, a GSK (glycogen synthase kinase) inhibitor, a HDAC (histone deacetylase) inhibitor, an IDO (indoleamine 2,3-dioxygenase) inhibitor, an IDH (isocitrate dehydrogenase, including IDH1) inhibitor, an IKK (1-Kappa-B kinase) inhibitor, a KDM5 (lysine demethylase) inhibitor, a LCK (lymphocyte-specific protein tyrosine kinase) inhibitor, a LOX (lysyl oxidase) inhibitor, a LOXL (lysyl oxidase like protein, including LOXL1, LOXL2, LOXL3, LOXL4, and/or LOXL5) inhibitor, a MTH (mut T homolog) inhibitor, a MEK (mitogen-activated protein kinase kinase) inhibitor, a matrix metalloprotease (MMP, including MMP2 and/or MMP9) inhibitor, a mitogen-activated protein kinases (MAPK) inhibitor, a PD-1 (programmed cell death protein 1) inhibitor, a PD-L1 (programmed death-ligand 1) inhibitor, a PDGF (platelet-derived growth factor) inhibitor, a phosphorylase kinase (PK) inhibitor, a PLK (polo-like kinase, including PLK1, 2, 3) inhibitor, a protein kinase (PK, including protein kinase A, B, C) inhibitor, a STK (serine/threonine kinase) inhibitor, a STAT (signal transduction and transcription) inhibitor, a serine/threonine-protein kinase inhibitor, a TBK (tank-binding kinase) inhibitor, a TLR (toll-like receptor modulators, including TLR-1, TLR-2, TLR-3, TLR-4, TLR-5, TLR-6, TLR-7, TLR-8, TLR-9, TLR-10, TLR-11, TLR-12, and/or TLR-13) inhibitor, a TK (tyrosine kinase) inhibitor, a TPL2 (serine/threonine kinase) inhibitor, a NEK9 inhibitor, an Abl inhibitor, a p38 kinase inhibitor, a PYK inhibitor, a PYK inhibitor, a c-Kit inhibitor, a NPM-ALK inhibitor, a Flt-3 inhibitor, a c-Met inhibitor, a KDR inhibitor, a TIE-2 inhibitor, a VEGFR inhibitor, a SRC inhibitor, a HCK inhibitor, a LYN inhibitor, a FYN inhibitor, a YES inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof. In some embodiments, the JAK inhibitor is N-(cyanomethyl)-4-[2-(4-morpholinoanilino)pyrimidin-4-yl]benzamide as named by ChemDraw (may also be referred to as CYT0387 or momelotinib) and may be synthesized by the methods described in U.S. Pat. No. 8,486,941. In certain embodiment, the SyK inhibitor is 6-(1H-indazol-6-yl)-N-(4-morpholinophenyl)imidazo[1,2-a]pyrazin-8-amine as named by ChemDraw (may also be referred to as 6-(1H-indazol-6-yl)-N-[4-(morpholin-4-yl)phenyl]imidazo[1,2-a]pyrazin-8-amine) and may be synthesized by the methods described in U.S. Pat. No. 8,450,321. In other embodiments, the BTK inhibitor is (S)-6-amino-9-(1-(but-2-ynoyl)pyrrolidin-3-yl)-7-(4-phenoxyphenyl)-7H-purin-8(9H)-one as named by ChemDraw (may also be 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one) and may be synthesized by the methods in U.S. Pat. No. 8,557,803.

Chemotherapeutic agents may be categorized by their mechanism of action into, for example, the following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (floxuridine, capecitabine, and cytarabine); purine analogs, folate antagonists and related inhibitors, antiproliferative/antimitotic agents including natural products such as vinca alkaloid (vinblastine, vincristine) and microtubule such as taxane (paclitaxel, docetaxel), vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide); DNA damaging agents (actinomycin, amsacrine, busulfan, carboplatin, chlorambucil, cisplatin, cyclophosphamide, Cytoxan, dactinomycin, daunorubicin, doxorubicin, epirubicin, iphosphamide, melphalan, merchlorehtamine, mitomycin, mitoxantrone, nitrosourea, procarbazine, taxol, taxotere, teniposide, etoposide, triethylenethiophosphoramide); antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents; antiproliferative/antimitotic alkylating agents such as nitrogen mustards cyclophosphamide and analogs, melphalan, chlorambucil), and (hexamethylmelamine and thiotepa), alkyl nitrosoureas (BCNU) and analogs, streptozocin), trazenes-dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes (cisplatin, oxiloplatinim, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones, hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole); anticoagulants (heparin, synthetic heparin salts and other inhibitors of thrombin); fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidine, clopidogrel; antimigratory agents; antisecretory agents (breveldin); immunosuppressives tacrolimus sirolimus azathioprine, mycophenolate; compounds (TNP-470, genistein) and growth factor inhibitors (vascular endothelial growth factor inhibitors, fibroblast growth factor inhibitors); angiotensin receptor blocker, nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab, rituximab); cell cycle inhibitors and differentiation inducers (tretinoin); inhibitors, topoisomerase inhibitors (doxorubicin (adriamycin), daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin, irinotecan and mitoxantrone, topotecan, irinotecan, camptothesin), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone); growth factor signal transduction kinase inhibitors; dysfunction inducers, toxins such as Cholera toxin, ricin, Pseudomonas exotoxin, Bordetella pertussis adenylate cyclase toxin, or diphtheria toxin, and caspase activators; and chromatin.

As used herein the term “chemotherapeutic agent” or “chemotherapeutic” (or “chemotherapy,” in the case of treatment with a chemotherapeutic agent) is meant to encompass any non-proteinaceous (i.e, non-peptidic) chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; emylerumines and memylamelamines including alfretamine, triemylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimemylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, foremustine, lomustine, nimustine, ranimustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammaII and calicheamicin phiI1, see, e.g., Agnew, Chem. Intl. Ed. Engl, 33:183-186 (1994); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carrninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as demopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogues such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replinisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; hestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformthine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; leucovorin; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; losoxantrone; fluoropyrimidine; folinic acid; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-tricUorotriemylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethane; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiopeta; taxoids, e.g., paclitaxel (TAXOL® and docetaxel (TAXOTERE®); chlorambucil; gemcitabine (Gemzar®); 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitroxantrone; vancristine; vinorelbine (Navelbine®); novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeoloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; FOLFIRI (fluorouracil, leucovorin, and irinotecan) and pharmaceutically acceptable salts, acids or derivatives of any of the above. One or more chemotherapeutic agent are used or included in the present application.

Also included in the definition of “chemotherapeutic agent” are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including Nolvadex™), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston®); inhibitors of the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate (Megace®), exemestane, formestane, fadrozole, vorozole (Rivisor®), letrozole (Femara®), and anastrozole (Arimidex®.); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprohde, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.

The anti-angiogenic agents include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATIN®, ENDOSTATIN®, suramin, squalamine, tissue inhibitor of metalloproteinase-1, tissue inhibitor of metalloproternase-2, plasminogen activator inhibitor-1, plasminogen activator inhibitor-2, cartilage-derived inhibitor, paclitaxel (nab-paclitaxel), platelet factor 4, protamine sulphate (clupeine), sulphated chitin derivatives (prepared from queen crab shells), sulphated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((1-azetidine-2-carboxylic acid (LACA), cishydroxyproline, d,I-3,4-dehydroproline, thiaproline, .alpha.-dipyridyl, beta-aminopropionitrile fumarate, 4-propyl-5-(4-pyridinyl)-2(3h)-oxazolone; methotrexate, mitoxantrone, heparin, interferons, 2 macroglobulin-serum, chimp-3, chymostatin, beta-cyclodextrin tetradecasulfate, eponemycin; fumagillin, gold sodium thiomalate, d-penicillamine (CDPT), beta-1-anticollagenase-serum, alpba-2-antiplasmin, bisantrene, lobenzarit disodium, n-2-carboxyphenyl-4-chloroanthronilic acid disodium or “CCA”, thalidomide; angiostatic steroid, cargboxynaminolmidazole; metalloproteinase inhibitors such as BB94. Other anti-angiogenesis agents include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: beta-FGF, alpha-FGF, FGF-5, VEGF isoforms, VEGF-C, HGF/SF and Ang-1/Ang-2. See Ferrara N. and Alitalo, K. “Clinical application of angiogenic growth factors and their inhibitors” (1999) Nature Medicine 5:1359-1364.

The anti-fibrotic agents include, but are not limited to, the compounds such as beta-aminoproprionitrile (BAPN), as well as the compounds disclosed in U.S. Pat. No. 4,965,288 to Palfreyman, et al., issued Oct. 23, 1990, entitled “Inhibitors of lysyl oxidase,” relating to inhibitors of lysyl oxidase and their use in the treatment of diseases and conditions associated with the abnormal deposition of collagen; U.S. Pat. No. 4,997,854 to Kagan, et al., issued Mar. 5, 1991, entitled “Anti-fibrotic agents and methods for inhibiting the activity of lysyl oxidase in situ using adjacently positioned diamine analogue substrate,” relating to compounds which inhibit LOX for the treatment of various pathological fibrotic states, which are herein incorporated by reference. Further exemplary inhibitors are described in U.S. Pat. No. 4,943,593 to Palfreyman, et al., issued Jul. 24, 1990, entitled “Inhibitors of lysyl oxidase,” relating to compounds such as 2-isobutyl-3-fluoro-, chloro-, or bromo-allylamine; as well as, e.g., U.S. Pat. Nos. 5,021,456; 5,5059,714; 5,120,764; 5,182,297; 5,252,608 (relating to 2-(1-naphthyloxymemyl)-3-fluoroallylamine); and U.S. Patent Application No. 2004/0248871, which are herein incorporated by reference. Exemplary anti-fibrotic agents also include the primary amines reacting with the carbonyl group of the active site of the lysyl oxidases, and more particularly those which produce, after binding with the carbonyl, a product stabilized by resonance, such as the following primary amines: emylenemamine, hydrazine, phenylhydrazine, and their derivatives, semicarbazide, and urea derivatives, aminonitriles, such as beta-aminopropionitrile (BAPN), or 2-nitroethylamine, unsaturated or saturated haloamines, such as 2-bromo-ethylamine, 2-chloroethylamine, 2-trifluoroethylamine, 3-bromopropylamine, p-halobenzylamines, selenohomocysteine lactone. Also, the anti-fibrotic agents are copper chelating agents, penetrating or not penetrating the cells. Exemplary compounds include indirect inhibitors such compounds blocking the aldehyde derivatives originating from the oxidative deamination of the lysyl and hydroxylysyl residues by the lysyl oxidases, such as the thiolamines, in particular D-penicillamine, or its analogues such as 2-amino-5-mercapto-5-methylhexanoic acid, D-2-amino-3-methyl-3-((2-acetamidoethyl)dithio)butanoic acid, p-2-amino-3-methyl-3-((2-aminoethyl)dithio)butanoic acid, sodium-4-((p-1-dimethyl-2-amino-2-carboxyethyl)dithio)butane sulphurate, 2-acetamidoethyl-2-acetamidoethanethiol sulphanate, sodium-4-mercaptobutanesulphinate trihydrate.

The immunotherapeutic agents include and are not limited to therapeutic antibodies suitable for treating patients; such as abagovomab, adecatumumab, afutuzumab, alemtuzumab, altumomab, amatuximab, anatumomab, arcitumomab, bavituximab, bectumomab, bevacizumab, bivatuzumab, blinatumomab, brentuximab, cantuzumab, catumaxomab, cetuximab, citatuzumab, cixutumumab, clivatuzumab, conatumumab, daratumumab, drozitumab, duligotumab, dusigitumab, detumomab, dacetuzumab, dalotuzumab, ecromeximab, elotuzumab, ensituximab, ertumaxomab, etaracizumab, farietuzumab, ficlatuzumab, figitumumab, flanvotumab, futuximab, ganitumab, gemtuzumab, girentuximab, glembatumumab, ibritumomab, igovomab, imgatuzumab, indatuximab, inotuzumab, intetumumab, ipilimumab, iratumumab, labetuzumab, lexatumumab, lintuzumab, lorvotuzumab, lucatumumab, mapatumumab, matuzumab, milatuzumab, minretumomab, mitumomab, moxetumomab, narnatumab, naptumomab, necitumumab, nimotuzumab, nofetumomabn, ocaratuzumab, ofatumumab, olaratumab, onartuzumab, oportuzumab, oregovomab, panitumumab, parsatuzumab, patritumab, pemtumomab, pertuzumab, pintumomab, pritumumab, racotumomab, radretumab, rilotumumab, rituximab, robatumumab, satumomab, sibrotuzumab, siltuximab, simtuzumab, solitomab, tacatuzumab, taplitumomab, tenatumomab, teprotumumab, tigatuzumab, tositumomab, trastuzumab, tucotuzumab, ublituximab, veltuzumab, vorsetuzumab, votumumab, zalutumumab, obinutuzumab, CC49 and 3F8. The exemplified therapeutic antibodies may be further labeled or combined with a radioisotope particle, such as indium In 111, yttrium Y 90, iodine I-131.

The application also provides method for treating a subject who is undergoing one or more standard therapies, such as chemotherapy, radiotherapy, immunotherapy, surgery, or combination thereof. Accordingly, one or more therapeutic agent or inhibitors may be administered before, during, or after administration of chemotherapy, radiotherapy, immunotherapy, surgery or combination thereof.

Other examples of chemotherapy treatments (including standard or experimental chemotherapies) are described below. In addition, treatment of certain lymphomas is reviewed in Cheson, B. D., Leonard, J. P., “Monoclonal Antibody Therapy for B-Cell Non-Hodgkin's Lymphoma” The New England Journal of Medicine 2008, 359(6), p. 613-626; and Wierda, W. G., “Current and Investigational Therapies for Patients with CLL” Hematology 2006, p. 285-294. Lymphoma incidence patterns in the United States is profiled in Morton, L. M., et al. “Lymphoma Incidence Patterns by WHO Subtype in the United States, 1992-2001” Blood 2006, 107(1), p. 265-276.

Examples of immunotherapeutic agents include, but are not limited to, rituximab (such as Rituxan), alemtuzumab (such as Campath, MabCampath), anti-CD19 antibodies, anti-CD20 antibodies, anti-MN-14 antibodies, anti-TRAIL, Anti-TRAIL DR4 and DR5 antibodies, anti-CD74 antibodies, apolizumab, bevacizumab, CHIR-12.12, epratuzumab (hLL2-anti-CD22 humanized antibody), galiximab, ha20, ibritumomab tiuxetan, lumiliximab, milatuzumab, ofatumumab, PRO131921, SGN-40, WT-1 analog peptide vaccine, WT1 126-134 peptide vaccine, tositumomab, autologous human tumor-derived HSPPC-96, and veltuzumab. Additional immunotherapy agents includes using cancer vaccines based upon the genetic makeup of an individual patient's tumor, such as lymphoma vaccine example is GTOP-99 (MyVax®).

Examples of chemotherapy agents include aldesleukin, alvocidib, antineoplaston AS2-1, antineoplaston A10, anti-thymocyte globulin, amifostine trihydrate, aminocamptothecin, arsenic trioxide, beta alethine, Bcl-2 family protein inhibitor ABT-263, ABT-199, BMS-345541, bortezomib (Velcade®), bryostatin 1, busulfan, carboplatin, campath-1H, CC-5103, carmustine, caspofungin acetate, clofarabine, cisplatin, Cladribine (Leustarin), Chlorambucil (Leukeran), Curcumin, cyclosporine, Cyclophosphamide (Cyloxan, Endoxan, Endoxana, Cyclostin), cytarabine, denileukin diftitox, dexamethasone, DT PACE, docetaxel, dolastatin 10, Doxorubicin (Adriamycin®, Adriblastine), doxorubicin hydrochloride, enzastaurin, epoetin alfa, etoposide, Everolimus (RAD001), fenretinide, filgrastim, melphalan, mesna, Flavopiridol, Fludarabine (Fludara), Geldanamycin (17-AAG), ifosfamide, irinotecan hydrochloride, ixabepilone, Lenalidomide (Revlimid®, CC-5013), lymphokine-activated killer cells, melphalan, methotrexate, mitoxantrone hydrochloride, motexafin gadolinium, mycophenolate mofetil, nelarabine, oblimersen (Genasense) Obatoclax (GX15-070), oblimersen, octreotide acetate, omega-3 fatty acids, oxaliplatin, paclitaxel, PD0332991, PEGylated liposomal doxorubicin hydrochloride, pegfilgrastim, Pentstatin (Nipent), perifosine, Prednisolone, Prednisone, R-roscovitine (Selicilib, CYC202), recombinant interferon alfa, recombinant interleukin-12, recombinant interleukin-11, recombinant flt3 ligand, recombinant human thrombopoietin, rituximab, sargramostim, sildenafil citrate, simvastatin, sirolimus, Styryl sulphones, tacrolimus, tanespimycin, Temsirolimus (CC1-779), Thalidomide, therapeutic allogeneic lymphocytes, thiotepa, tipifarnib, Velcade® (bortezomib or PS-341), Vincristine (Oncovin), vincristine sulfate, vinorelbine ditartrate, Vorinostat (SAHA), vorinostat, and FR (fludarabine, rituximab), CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), CVP (cyclophosphamide, vincristine and prednisone), FCM (fludarabine, cyclophosphamide, mitoxantrone), FCR (fludarabine, cyclophosphamide, rituximab), hyperCVAD (hyperfractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate, cytarabine), ICE (iphosphamide, carboplatin and etoposide), MCP (mitoxantrone, chlorambucil, and prednisolone), R-CHOP (rituximab plus CHOP), R-CVP (rituximab plus CVP), R-FCM (rituximab plus FCM), R-ICE (rituximab-ICE), and R-MCP (R-MCP).

The therapeutic treatments can be supplemented or combined with any of the abovementioned therapies with stem cell transplantation or treatment. One example of modified approach is radioimmunotherapy, wherein a monoclonal antibody is combined with a radioisotope particle, such as indium In 111, yttrium Y 90, iodine I-131. Examples of combination therapies include, but are not limited to, Iodine-131 tositumomab (Bexxar®), Yttrium-90 ibritumomab tiuxetan (Zevalin®), Bexxar® with CHOP.

Other therapeutic procedures include peripheral blood stem cell transplantation, autologous hematopoietic stem cell transplantation, autologous bone marrow transplantation, antibody therapy, biological therapy, enzyme inhibitor therapy, total body irradiation, infusion of stem cells, bone marrow ablation with stem cell support, in vitro-treated peripheral blood stem cell transplantation, umbilical cord blood transplantation, immunoenzyme technique, pharmacological study, low-LET cobalt-60 gamma ray therapy, bleomycin, conventional surgery, radiation therapy, and nonmyeloablative allogeneic hematopoietic stem cell transplantation.

In some embodiments, the methods include administering a compound of the formula described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, in a therapeutically effective amount to a human in need thereof. The method can be employed to treat a patient who has or is believed to have a disease or condition whose symptoms or pathology is mediated by expression or activity of PI3Kβ and/or PI3Kδ. The patient may be a mammal or a human. In certain embodiment, the patient may be a human.

“Treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired clinical results may include one or more of the following: a) inhibiting the disease or condition (e.g., decreasing one or more symptoms resulting from the disease or condition, and/or diminishing the extent of the disease or condition); b) slowing or arresting the development of one or more clinical symptoms associated with the disease or condition (e.g., stabilizing the disease or condition, preventing or delaying the worsening or progression of the disease or condition, and/or preventing or delaying the spread (e.g., metastasis) of the disease or condition); and/or c) relieving the disease, that is, causing the regression of clinical symptoms (e.g., ameliorating the disease state, providing partial or total remission of the disease or condition, enhancing the effect of another medication, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival.

“Prevention” or “preventing” mean any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop. Compounds may, in some embodiments, be administered to a subject (including a human) who is at risk or has a family history of the disease or condition.

“Subject” or “patient” refer to an animal, such as a mammal (including a human), that has been or will be the object of treatment, observation or experiment. The methods described herein may be useful in human therapy and/or veterinary applications. In some embodiments, the subject is a mammal. In one embodiment, the subject is a human. “Human in need thereof” refers to a human who may have or is suspect to have diseases, or disorders, or conditions that would benefit from certain treatment; for example, being treated with the PI3K inhibitor of the compounds according to the present application. In certain embodiments, the subject may be a human who (i) has not received any treatment including chemotherapy treatment, (ii) is substantially refractory to at least one chemotherapy treatment, (iii) is in relapse after treatment with chemotherapy, or both (i) and (ii). In some of embodiments, the subject is refractory to at least one, at least two, at least three, or at least four chemotherapy treatments (including standard or experimental chemotherapies).

The terms “therapeutically effective amount” or “effective amount” of a compound of the present application or a pharmaceutically acceptable salt, isomers, prodrug, or solvate thereof, mean an amount sufficient to effect treatment when administered to a subject, to provide a therapeutic benefit such as amelioration of symptoms or slowing of disease progression. For example, a therapeutically effective amount may be an amount sufficient to decrease a symptom of a disease or condition responsive to inhibition of PI3Kδ and PI3Kβ activity. The therapeutically effective amount may vary depending on the subject, and disease or condition being treated, the weight and age of the subject, the severity of the disease or condition, and the manner of administering, which can readily be determined by one or ordinary skill in the art.

In addition to the therapeutic uses, the compounds described herein have the selectivity or selective inhibition to certain PI3K isoforms. In one embodiment, the compounds have selectivity to PI3Kβ. In some embodiments, the compounds have selectivity to PI3Kδ. In yet other embodiments, the compounds have selectivity to PI3Kβ and PI3Kδ. The selectivity to PI3K isoforms may be determined by measuring the compound's activity in inhibiting certain PI3K isoforms using the assay described in the example below or the methods commonly used. It is understood that the conditions (e.g. the reagent concentration or the incubation temperature) may be varied and the results of the assay may vary. In some instances, the value may vary within a range of one to three-folds.

The term “inhibition” indicates a decrease in the baseline activity of a biological activity or process. The term “inhibition of activity of PI3K isoforms” or variants thereof refer to a decrease in activity in any PI3K isoform (e.g., alpha, beta, gamma, or delta) as a direct or indirect response to the presence of a compound of any of the formula described herein relative to the activity of PI3K isoform in the absence of such compound. “Inhibition of PI3Kδ and/or PI3Kβ activities” or variants thereof refer to a decrease in PI3Kδ and/or PI3Kβ activities as a direct or indirect response to the presence of the compounds described herein, relative to the activities of PI3Kδ and/or PI3Kβ in the absence of such compound. In some embodiments, the inhibition of PI3K isoform activities may be compared in the same subject prior to treatment, or other subjects not receiving the treatment.

Without being bound to any theory, the decrease in the activity of PI3K may be due to the direct interaction of the compound with PI3K, or due to the interaction of the compounds described herein with one or more other factors that affect PI3K activity. For example, the presence of the compounds may decrease the activities of PI3Kδ and/or PI3Kβ by directly binding to PI3Kδ and/or PI3Kβ, by causing (directly or indirectly) another factor to decrease PI3Kδ and/or PI3Kβ activities, or by (directly or indirectly) decreasing the amount of PI3Kδ and/or PI3Kβ present in the cell or organism.

The term “PI3K inhibitor” or variant thereof refers to a compound that inhibits the activity of PI3K. The term “PI3K isoform selective inhibitor” or variant thereof refers to a compound that inhibits the activity of one or more PI3K isoforms more effectively than the other remaining PI3K isoforms. By way of example, the term “PI3Kβ selective inhibitor” generally refers to a compound that inhibits the activity of the PI3Kβ isoform more effectively than other isoforms of the PI3K family, and the term “PI3Kδ selective inhibitor” generally refers to a compound that inhibits the activity of the PI3Kδ isoform more effectively than other isoforms of the PI3K family. The term “dual PI3Kδ/β selective inhibitor” generally refers to a compound that inhibits the activity of both PI3Kδ and PI3Kβ isoforms more effectively than other isoforms of the PI3K family (e.g., PI3Kα or γ).

The relative efficacies of compounds as inhibitors of an enzyme activity (or other biological activity) can be established by determining the concentrations at which each compound inhibits the activity to a predefined extent and then comparing the results. In one embodiment, the efficacy of a compound as an inhibitor of one or more PI3K isoforms can be measured by the compound concentration that inhibits 50% of the activity in a biochemical assay, i.e., the 50% inhibitory concentration or “IC50”. The determination of IC50 values can be accomplished using conventional techniques known in the art, including the techniques described in the Examples below. In general, an IC50 can be determined by measuring the activity of a given enzyme in the presence of a range of concentrations of the compound under the study. The experimentally obtained values of enzyme activity may then be plotted against the compound concentrations used. The concentration of the inhibitor that shows 50% enzyme activity (as compared to the activity in the absence of any inhibitor) is taken as the IC50 value. Analogously, other inhibitory concentrations can be defined through appropriate determinations of activity. For example, in some settings it may be desirable to establish a 90% inhibitory concentration, i.e., IC90.

According to the present application, a PI3Kβ selective inhibitor is a compound that exhibits a 50% inhibitory concentration (IC50) with respect to PI3Kβ that is at least 10-fold, at least 20-fold, at least 30-fold, at least 50-fold, at least 100-fold, at least 200-fold, or at least 500-fold lower than the IC50 with respect to either PI3Kα or PI3Kγ or both PI3Kα and PI3Kγ. In addition, a PI3Kδ/β selective inhibitor is a compound that exhibits a 50% inhibitory concentration (IC50) with respect to PI3Kβ and PI3Kδ that is at least 10-fold, at least 20-fold, at least 30-fold, at least 50-fold, at least 75-fold, at least 100-fold, at least 200-fold, and at least 500-fold lower than the IC50 with respect to either PI3Kα or PI3Kγ. The dual PI3Kδ/β selective inhibitor may have the same or similar IC50 to both PI3Kδ and PI3Kβ or may have different IC50 to either PI3Kδ or PI3Kβ. As used herein, the term “potency,” “potent,” or variants thereof refer to the compound exhibiting an IC50 value that is less than 100 nM. When comparing two compounds, the compound that exhibits a lower IC50 value is referred to as a more potent inhibitor.

The compounds of the present application exhibit unexpected selectivity to PI3Kβ. As shown in the example, certain compounds in Table 1 exhibit low IC50 values (e.g. 1 to 100 nM) to both PI3Kβ and PI3Kδ. Certain compounds in Table 1a also exhibited such activity to PI3K isoforms. Also, certain compounds of formula (I) exhibited at least between 10-fold to 400-fold lower IC50 values for PI3Kβ than PI3Kγ, suggesting the compounds exhibit selectivity to PI3Kβ compared to PI3Kγ (i.e., inhibits the activity of the PI3Kβ isoform more effectively than the PI3Kγ isoform as shown by the PI3Kγ/PI3Kβ ratio). Some compounds of formula (IV) showed similar selectivity. Moreover, the compounds described herein exhibit selectivity to both PI3Kβ and PI3Kδ. The compound of (S)-2,4-diamino-6-((1-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile, described in U.S. Provisional Application No. 61/745,437, exhibited less selectivity to PI3Kγ (e.g. the PI3Kγ/PI3Kβ ratio is less than 1-fold). The results of the present application suggest that the compounds described herein are dual selective inhibitors of PI3Kδ and PI3Kβ and exhibit selectivity to PI3Kβ compared to PI3Kγ. Each of the patents and the patent publications provided in the present application is hereby incorporated by reference in the entirety.

The methods described herein may be applied to cell populations in vivo or ex vivo. “In vivo” means within a living individual, as within an animal or human. In this context, the methods described herein may be used therapeutically in an individual. “Ex vivo” means outside of a living individual. Examples of ex vivo cell populations include in vitro cell cultures and biological samples including fluid or tissue samples obtained from individuals. Such samples may be obtained by methods well known in the art. Exemplary biological fluid samples include blood, cerebrospinal fluid, urine, and saliva. Exemplary tissue samples include tumors and biopsies thereof. In this context, the compounds may be used for a variety of purposes, including therapeutic and experimental purposes. For example, it may be used ex vivo to determine the optimal schedule and/or dosing of administration of a PI3K selective inhibitor for a given indication, cell type, individual, and other parameters. Information gleaned from such use may be used for experimental purposes or in the clinic to set protocols for in vivo treatment. Other ex vivo uses for which the invention may be suited are described below or will become apparent to those skilled in the art. The compounds of the formula described herein or a pharmaceutically acceptable salt, prodrug, or solvate thereof, may be further characterized to examine the safety or tolerance dosage in human or non-human subjects. Such properties may be examined using commonly known methods to those skilled in the art.

Compared to other PI3K isoforms, PI3Kδ is generally expressed in hematopoietic cells. Also, PI3Kβ is generally mis-regulated in certain cancer cells. Aberrant proliferation of cells often interferes with normal tissue function, which may result in abnormal cellular response such as immunity, inflammation, and/or apoptosis. The selective inhibitors to PI3Kδ and/or PI3Kβ are useful in treating, inhibiting, or preventing aberrant proliferation of cancerous and/or hematopoietic cells and ameliorating the symptoms and secondary conditions.

The compounds described herein may be used to treat subjects having various disease states, disorders, and conditions (also collectively referred to as “indications”) associated with PI3K isoforms or their activities. As used herein, the terms “diseases,” “disorders,” “conditions” are used interchangeably. Such indications may include, for example, cancer, including hematologic malignancies (e.g. leukemias and lymphomas, myeloproliferative disorders, myelodysplastic syndromes, plasma cell neoplasms) and solid tumors, inflammation, fibrosis, allergic conditions (including hypersensitivity), cardiovascular diseases, neurodegenerative diseases, renal disorders, viral infections, obesity, and autoimmune diseases.

In other embodiments, the compounds described herein may be used to treat cancers that are mediated by, dependent on, or associated with PI3K activity. In certain embodiments, the disease or condition is an autoimmune disease, an inflammatory disease, or a cancer. In some embodiments, the disease or condition is chosen from rheumatoid arthritis, osteoarthritis, atherosclerosis, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, asthma, chronic obstructive airways disease, pneumonitis, dermatitis, alopecia, nephritis, vasculitis, atherosclerosis, Alzheimer's disease, hepatitis, primary biliary cirrhosis, sclerosing cholangitis, diabetes (including type I diabetes), acute rejection of transplanted organs, lymphomas, multiple myelomas, leukemias, neoplasms and solid tumors.

In other embodiments, the disease is a solid tumor. By way of examples, the solid tumor includes but is not limited to pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, rectum cancer, liver cancer, kidney cancer, stomach cancer, skin cancer, gastric cancer, esophageal cancer, head and neck cancer, melanoma, neuroendocrine cancers, CNS cancers (e.g., neuroblastoma), brain tumors (e.g., glioma, anaplastic oligodendroglioma, adult glioblastoma multiforme, and adult anaplastic astrocytoma), bone cancer, or soft tissue sarcoma. In some embodiments, the solid tumor is non-small cell lung cancer, small-cell lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, pancreatic cancer, prostate cancer, or breast cancer.

The present application also provides a method for treating a human in need thereof, who has or is suspected of having a disease or condition responsive or believed to be responsive to the inhibition of PI3Kδ and/or PI3Kβ activity by administering to the subject a compound of the formulae described herein or a pharmaceutically acceptable salt, enantiomer, atropisomer, tautomer, prodrug, or solvate thereof.

Additionally, the application provides a method of inhibiting kinase activity of a PI3Kδ and/or PI3Kβ polypeptides by contacting the polypeptides with a compound of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof.

Moreover, the application provides a method of decreasing cell viability, increasing cell death or apoptosis, increasing interference with PI3K signaling pathways (including AKT, S6RP, ERK phosphorylation), and/or reduction in chemokine production with an effective amount of a compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof.

The application further provides a method of disrupting leukocyte function comprising contacting the leukocytes with an effective amount of a compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof, in a human in need thereof.

Provided is also a method of inhibiting growth or proliferation of cancer cells comprising contacting the cancer cells with an effective amount of a compound of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof.

Kits

Provided herein are also kits that include a compound of the formulae of the present application or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, and suitable packaging. In one embodiment, a kit further includes instructions for use. In one aspect, a kit includes a compound of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, and a label and/or instructions for use of the compounds in the treatment of the indications, including the diseases or conditions, described herein.

Provided herein are also articles of manufacture that include a compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, in a suitable container. The container may be a vial, jar, ampoule, preloaded syringe, and intravenous bag.

Pharmaceutical Compositions and Modes of Administration

Compounds provided herein are usually administered in the form of pharmaceutical compositions. Thus, provides herein are also pharmaceutical compositions that contain one or more of the compounds of any of the formulae disclosed herein or a pharmaceutically acceptable salt, isomers, prodrug, or solvate thereof, and one or more pharmaceutically acceptable vehicles selected from carriers, adjuvants and excipients. Suitable pharmaceutically acceptable vehicles may include, for example, inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants. Such compositions are prepared in a manner well known in the pharmaceutical art. See, e.g., Remington's Pharmaceutical Sciences, Mace Publishing Co., Philadelphia, Pa. 17th Ed. (1985); and Modern Pharmaceutics, Marcel Dekker, Inc. 3rd Ed. (G. S. Banker & C. T. Rhodes, Eds.).

The pharmaceutical compositions may be administered in either single or multiple doses. The pharmaceutical composition may be administered by various methods including, for example, rectal, buccal, intranasal, and transdermal routes. In certain embodiments, the pharmaceutical composition may be administered by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant. In some embodiments, the pharmaceutical composition is administered orally.

One mode for administration is parenteral, for example, by injection. The forms in which the pharmaceutical compositions described herein may be incorporated for administration by injection include, for example, aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.

Oral administration may be another route for administration of the compounds described herein. Administration may be via, for example, capsule or enteric coated tablets. In making the pharmaceutical compositions that include at least one compound of any of the formulae described herein or a pharmaceutically acceptable salt, prodrug, or solvate thereof, the active ingredient is usually diluted by an excipient and/or enclosed within such a carrier that can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be in the form of a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders. In certain embodiments, the pharmaceutical composition is in the form of tablets.

Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose. The formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl and propylhydroxy-benzoates; sweetening agents; and flavoring agents.

The compositions that include at least one compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the subject by employing procedures known in the art. Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolutional systems containing polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Pat. Nos. 3,845,770; 4,326,525; 4,902,514; and 5,616,345. Another formulation for use in the methods of the present invention employs transdermal delivery devices (“patches”). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds described herein in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.

For preparing solid compositions such as tablets, the principal active ingredient may be mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of any of the above formulae or a pharmaceutically acceptable salt, prodrug, or solvate thereof. When referring to these preformulation compositions as homogeneous, the active ingredient may be dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.

The tablets or pills of the compounds described herein may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action, or to protect from the acid conditions of the stomach. For example, the tablet or pill can include an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.

Compositions for inhalation or insufflation may include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. In other embodiments, compositions in pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a facemask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.

Dosing

The specific dose level of a compound of the formulae described herein for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease in the subject undergoing therapy. For example, a dosage may be expressed as a number of milligrams of a compound of the formula per kilogram of the subject's body weight (mg/kg). Dosages of between about 0.01 and 200 mg/kg may be appropriate. In some embodiments, about 0.01 and 150 mg/kg may be appropriate. In other embodiments a dosage of between 0.05 and 100 mg/kg may be appropriate. Normalizing according to the subject's body weight is particularly useful when adjusting dosages between subjects of widely disparate size, such as occurs when using the drug in both children and adult humans or when converting an effective dosage in a non-human subject such as dog to a dosage suitable for a human subject.

The daily dosage may also be described as a total amount of a compound of the formulae administered per dose or per day. Daily dosage of a compound may be between about 1 mg and 2,000 mg, between about 1,000 to 2,000 mg/day, between about 1 to 1,000 mg/day, between about 1 to 500 mg/day, between about 100 to 150 mg/day, between about 1 to 100 mg/day, between about between about 1 to 50 mg/day, between about 50 to 100 mg/day, between about 100 to 125 mg/day, between about 100 to 150 mg/day, between about 100 to 175 mg/day, between about 100 to 200 mg/day, between about 100 to 225 mg/day, between about 100 to 250 mg/day, between about 100 to 350 mg/day, between about 100 to 400 mg/day, between about 100 to 450 mg/day, or between about 100 to 500 mg/day.

When administered orally, the total daily dosage for a human subject may be between 1 mg and 1,000 mg/day, between about 1 to 100 mg/day, between about 1 to 50 mg/day, between about 50 to 100 mg/day, between 100 to 200 mg/day, between about 200 to 300 mg/day, between about 300 to 400 mg/day, between about 400 to 500 mg/day, between about 100 to 150 mg/day, between about 150 to 200 mg/day, between about 200 to 250 mg/day, between about 75 to 150 mg/day, or between about 150 to 300 mg/day.

The compounds of the present application or the compositions thereof may be administered once, twice, three, or four times daily, using any suitable mode described above. Also, administration or treatment with the compounds according to any of the formulae described herein may be continued for a number of days; for example, commonly treatment would continue for at least 7 days, 14 days, or 28 days, for one cycle of treatment. In some treatment, the compound or the composition thereof is administered continuously, i.e. every day. Treatment cycles are well known in cancer chemotherapy, and are frequently alternated with resting periods of about 1 to 28 days, commonly about 7 days or about 14 days, between cycles. The treatment cycles, in other embodiments, may also be continuous.

In a particular embodiment, the method comprises administering to the subject an initial daily dose of about 1 to 500 mg of a compound of the above formula and increasing the dose by increments until clinical efficacy is achieved. Increments of about 1, 5, 10, 25, 50, 75, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.

Synthesis of the Compounds

The compounds of the present application may be prepared using the methods disclosed herein and routine modifications thereof, which will be apparent given the disclosure herein and methods well known in the art. Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of typical compounds described herein may be accomplished as described in the following examples. If available, reagents may be purchased commercially, e.g., from Sigma Aldrich or other chemical suppliers. In general, compounds described herein are typically stable and isolatable at room temperature and pressure.

General Synthesis

Typical embodiments of compounds described herein may be synthesized using the general reaction schemes described below. It will be apparent given the description herein that the general schemes may be altered by substitution of the starting materials with other materials having similar structures to result in products that are correspondingly different. Descriptions of syntheses follow to provide numerous examples of how the starting materials may vary to provide corresponding products. Given a desired product for which the substituent groups are defined, the necessary starting materials generally may be determined by inspection. Starting materials are typically obtained from commercial sources or synthesized using published methods. For synthesizing compounds which are embodiments described in the present disclosure, inspection of the structure of the compound to be synthesized will provide the identity of each substituent group. The identity of the final product will generally render apparent the identity of the necessary starting materials by a simple process of inspection, given the examples herein.

Synthetic Reaction Parameters

The terms “solvent”, “inert organic solvent”, or “inert solvent” refer to a solvent inert under the conditions of the reaction being described in conjunction therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran (“THF”), dimethylformamide (“DMF”), chloroform, methylene chloride (or dichloromethane), diethyl ether, methanol, and the like. Unless specified to the contrary, the solvents used in the reactions of the present invention are inert organic solvents, and the reactions are carried out under an inert gas, preferably nitrogen.

The compounds of formula (I) may be prepared using the method shown in Reaction Scheme I.

Step 1—Preparation of a Compound of Formula (1)

The compound of formula (1) can be made by combining compounds (A), (B) and (C) in the presence of a dehydrating agent. PG may be a protecting group such as t-butoxycarbonyl (Boc) or fluorenylmethoxycarbonyl (Fmoc). Compounds (A), (B) and (C) are commercially available or can be made by the methods that are commonly known or used by one skilled in the art. R1, R5, R′, and R3 are defined as in the formula (I). Ra is either a protecting group such as NHBoc or R2 as defined for the formula (I). Compound (A) is mixed with Compound (B) in the presence of a coupling agent such as diphenyl phosphite in a solvent such as pyridine. After stirring at a temperature between ambient and 100° C. for 1 to 5 hours, compound (C) is added. The mixture is further stirred at a temperature between ambient and 100° C. for 5 to 24 hours and cooled to room temperature. To extract the compound of formula (1), an organic solvent such as ethyl acetate (EtOAc) is added. Then the reaction is washed with mild acid, water, and brine. The organic phase is concentrated to obtain the compound of formula (1). The compound of formula (1) is purified by any suitable methods known in the art, such as chromatography on silica gel. Alternatively, the compound of formula (1) is purified directly without an aqueous work-up. Alternatively, the compound of formula (1) is used in the next step without purification. In cases where it is desired to convert a nitrile to a carboxamide, reaction under conditions known to those skilled in the art may be used such as treatment with Parkins-Ghaffar catalyst. In cases where a vicinal amino alcohol is desired for —B—Ra, an olefin can be reacted with an appropriate oxidant, such as m-chloroperbenzoic acid (mCPBA) to give the epoxide, which can be subsequently reacted with an excess of the amine, R2NH2, in the presence of an additive, such as lithium perchlorate, to give the amino alcohol. In cases where a sulfone is desired for —B—R2, a sulfide can be reacted with an appropriate oxidant, such as m-chloroperbenzoic acid (mCPBA) or hydrogen peroxide to give the sulfone.

Step 2—Preparation of a Compound of Formula (2)

The compound of formula (2) can be made by removing the protecting group(s) from the compound of formula (1). Different conditions will be used depending on the protecting group being utilized. If it is desired to remove a Boc group, the compound of formula (1) is dissolved in a suitable solvent and treated with a suitable acid. By way of example, suitable solvents include dichloromethane or dioxane, and suitable acids include trifluoroacetic acid, hydrochloric acid, or boron tribromide (BBr3). The reaction is carried out at temperatures between −78° C. to ambient temperature. After the reaction is complete, solvent is removed to obtain the compound of formula (2). If it is desired to remove an Fmoc group the compound of formula (1) is dissolved in a suitable solvent and treated with a suitable base. By way of example, suitable solvents include dichloromethane or DMF, and suitable bases include piperidine. The reaction is carried out at temperatures between 0° C. to 50° C. for between 0.5 and 24 hours. After the reaction is complete, the reaction may be diluted with water and a suitable solvent for extraction such as EtOAc or dichloromethane. The solvent is removed to obtain the compound of formula (2) and used without further purification or purified via methods known, such as chromatography.

Step 3—Preparation of Compounds of Formulae (I) and (3)

The compound of formula (I) where A is a single bond and Ra is an R2, can generally be prepared by coupling compound of formula (2) and an appropriately substituted R4—X, where X is a halo, in the presence of a suitable base in a suitable solvent. An example of a suitable base is diisopropylethylamine, triethylamine or N-methylmorpholine. An example of a suitable solvent is N-methylpyrrolidone (NMP), DMF, DMSO, or isopropanol. Also, an additive such as potassium fluoride may be used. The reaction is typically performed at a temperature between 50° C. to 150° C. for about 30 minutes to 24 hours. Alternatively the reaction can be performed in a microwave at a temperature between 100° C. to 150° C. for about 30 minutes to 24 hours. The compound of formula (I) where A is C(O) and Ra is an R2, may generally be prepared by coupling a compound of formula (2) with a suitable acid in the presence of a coupling agent, such as HATU, DCC, or carbonyl diimidazole in the presence of a base such as triethylamine or diisopropylethylamine in a suitable solvent such as dichloromethane or THF. Water can be added to quench the reaction upon completion, and the precipitate may be filtered then dissolved in an organic solvent such as dichloromethane (DCM). Alternatively, the reaction can be diluted with an appropriate solvent such as EtOAc or dichloromethane instead of filtering off the solid. The product can be isolated by methods known in the art, for example by removal of solvent under reduced pressure. The product can be purified using any suitable methods known in the art, for example, recrystallization, precipitation, or chromatography of the residue. For the compound of formula (3) where Ra is NR2xBoc, the Boc group may be removed by methods known to one of skill in the art and described above in step 2 to give a compound of formula (I), where R2 is NHR2x If further functionalization is desired, a compound of formula (I) where R2 is NHR2x can be converted to an amide, R2 is NR2xC(O)R2y by methods known to one of skill in the art, such as reaction with an acid, HOC(O)R2y, and a coupling agent, such as HATU, as described above. Furthermore, a compound of formula (I), where R2 is NHR2x may be alkylated by methods known to one of skill in the art through the reaction of the amine with a compound containing an aldehyde or ketone in the presence of a reducing agent, such as NaCNBH3 or sodium triacetoxyborohydride, in a suitable solvent such as methanol or dichloromethane to give a compound of formula (I), where R2 is NR2xR2y. Furthermore, a compound of formula (I), where R2 is NHR2x may be arylated or heteroarylated by methods known to one of skill such as by reaction of the amine with an activated aromatic or heteroaromatic ring in the presence of a base, such as triethylamine or diisopropylethylamine, in a suitable solvent such as DMF at a temperature between 50° C. to 150° C. to give a compound of formula (I), where R2 is NR2xR2y. Furthermore, a compound of formula (I) where R2 is CN may be converted to a compound of formula (I) where R2 is CONH2 by reaction under conditions known to those skilled in the art such as treatment with Parkins-Ghaffar catalyst. Furthermore, a compound of formula (I) where R2 is OH may be prepared by deprotecting a compound of formula (3) where Ra is OPG, where PG is an appropriate protecting group. Appropriate protecting groups may be acetate, benzoate and the like. Such protecting groups may be removed by treatment with reagents known to those skilled in the art such as LiOH or NaOH.

The compounds of formula (I) may also be prepared using the method shown in Reaction Scheme II.

Preparation of a Compound of Formula (4)

The compound of formula (4) can be made by removing the protecting group(s) from the compound of formula (1) wherein Ra is NR2y; and PG is Fmoc. The compound of formula (1) wherein Ra is NR2y; and PG is Fmoc is dissolved in a suitable solvent and treated with a suitable acid. By way of example, suitable solvents include dichloromethane or dioxane, and suitable acids include trifluoroacetic acid, hydrochloric acid, or boron tribromide (BBr3). The reaction is carried out at temperatures between −78° C. to ambient temperature. After the reaction is complete, solvent is removed to obtain the compound of formula (4).

Preparation of a Compound of Formula (5)

The compound of formula (5) can be made by methods known to one of skill in the art, such as reaction of the compound of formula (4) and an acid with a coupling agent, such as HATU, as described above in step 3. Furthermore, the compound of formula (4) may be alkylated by methods known to one of skill in the art through the reaction with a compound containing an aldehyde or ketone, in the presence of a reducing agent, such as NaCNBH3 or sodium triacetoxyborohydride, in a suitable solvent such as methanol or dichloromethane as described above in step 3.

Preparation of a Compound of Formula (6)

The compound of formula (6) can be made by removing the Fmoc from the compound of formula (5) as described in step 2. The compound of formula (5) is dissolved in a suitable solvent and treated with a suitable base. By way of example, suitable solvents include dichloromethane or DMF, and suitable bases include piperidine. The reaction is carried out at temperatures between 0° C. to 50° C. for between 0.5 and 24 hours. After the reaction is complete, the reaction may be diluted with water and a suitable solvent for extraction such as EtOAc or dichloromethane. The solvent is removed to obtain the compound of formula (6) and used without further purification or purified via methods known, such as chromatography. It should be understood that the compounds of formula (I) can be prepared according to the methods provided in Reaction Scheme 1 or 2, starting from materials known to one of skill in the art. After synthesis, the compounds may be isolated in the form of a free base or a trifluoroacetic acid salt and further characterized by NMR. The resulting compounds and their NMR characterizations may represent either the free base or salt form. The ratio of parent compound and corresponding salt is not determined.

Example 1 Preparation of a Compound of Formula (1)

A. Preparation of a compound of formula (1) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, Ra is NHBoc, R5 is hydrogen, PG is Fmoc, and R3 is phenyl

To a solution of 6-chloroanthranlic acid (0.92 g, 5.4 mmol) and Fmoc-L-phenylglycine (2.0 g, 5.4 mmol) in pyridine (2.7 mL) was added diphenyl phosphite (3.6 mL, 19 mmol). The mixture stirred at 40° C. for 1 h. N-Boc-1,3-propanediamine (0.95 g, 5.4 mmol) was added and the reaction mixture stirred overnight at 55° C. The mixture was diluted with EtOAc, washed with 1 N HCl (aq), sat bicarbonate solution, and brine. The organic layer was dried (Na2SO4), filtered, and concentrated onto silica gel. The crude product mixture was purified via flash chromatography (SiO2, 0-55% EtOAc/hexanes) to give (S)-tert-butyl (3-(2-(((((9H-fluoren-9-yl)methoxy)carbonyl)amino)(phenyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)(methyl)carbamate. ES/MS 665.3 (M+H+).

B. Preparation of the below compound of formula (1), using the methods described in Example 1A and Reaction Scheme I

  • (S)-tert-butyl (4-(2-(1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)-2-methylbutan-2-yl)carbamate;
  • (S)-tert-butyl (2-((2-(1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)(benzyl)amino)ethyl)carbamate;
  • (S)-tert-butyl (2-((2-(1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)(methyl)amino)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-morpholinopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • tert-butyl (2-(2-((S)-1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (1-(3-(but-3-en-1-yl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • tert-butyl (3-(2-((S)-1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl)carbamate;
  • tert-butyl (3-(2-((S)-1-(((9H-fluoren-9-yl)methoxy)carbonylamino)-5-chloro-4-oxoquinazolin-3(4H)-yl)-2-methylpropyl)carbamate;
  • (S)-tert-butyl (4-(2-(1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl)carbamate;
  • tert-butyl (3-(2-((1S)-1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-5-methyl-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-(dimethylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(2-(dimethylamino)ethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • tert-butyl (3-(5-chloro-2-((1S)-1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • tert-butyl (3-(5-chloro-2-((1S)-1-(((9H-fluoren-9-yl)methoxy)carbonylamino)ethyl)-4-oxoquinazolin-3(4H)-yl)propyl)(methyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-methylbutyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl ((5-chloro-3-(2-cyanoethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl ((5-chloro-3-(4-cyanobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)(methyl)carbamate;
  • 9H-fluoren-9-ylmethyl N-[(1S)-1-[3-[3-(tert-butoxycarbonylamino)propyl]-5-chloro-4-oxo-quinazolin-2-yl]propyl]carbamate;
  • tert-butyl N-[(1S)-1-[3-[3-[benzyl(methyl)amino]propyl]-5-chloro-4-oxo-quinazolin-2-yl]ethyl]carbamate;
  • 9H-fluoren-9-ylmethyl N—[(S)-[3-[3-(tert-butoxycarbonylamino)propyl]-5-chloro-4-oxo-quinazolin-2-yl]-cyclopropyl-methyl]carbamate;
  • tert-butyl N—[(S)-[5-chloro-3-(3-cyanopropyl)-4-oxo-quinazolin-2-yl]-cyclopropyl-methyl]carbamate;
  • tert-butyl N-[3-[5-chloro-2-[(1S)-1-(9H-fluoren-9-ylmethoxycarbonylamino)ethyl]-4-oxo-quinazolin-3-yl]propyl]-N-methyl-carbamate;
  • 9H-fluoren-9-ylmethyl N-[(1S)-1-[3-[3-(tert-butoxycarbonylamino)propyl]-5-chloro-4-oxo-quinazolin-2-yl]ethyl]carbamate;
  • tert-butyl N—[(S)-cyclopropyl-[5,8-dichloro-3-(3-cyanopropyl)-4-oxo-quinazolin-2-yl]methyl]carbamate;
  • tert-butyl N-[3-[5-chloro-2-[(S)-cyclopropyl-(9H-fluoren-9-ylmethoxycarbonylamino)methyl]-4-oxo-quinazolin-3-yl]propyl]-N-methyl-carbamate;
  • (S)-tert-butyl (1-(3-((1H-pyrazol-3-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(3-((1H-pyrazol-4-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(3-((1H-imidazol-2-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(3-((1H-1,2,4-triazol-5-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (1R,3s)-3-(2-((S)-1-((tert-butoxycarbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (S)-3-(2-(1-((tert-butoxycarbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl benzoate;
  • 3-(2-((S)-1-((tert-butoxycarbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl benzoate;
  • (1S,4r)-4-(2-((S)-1-((tert-butoxycarbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclohexyl benzoate;
  • (S)-tert-butyl 4-((2-(1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)morpholine-4-carboxylate;
  • (S)-tert-butyl (3-(2-(1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl)carbamate;
  • tert-butyl 3-((2-((S)-1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)pyrrolidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)pyrrolidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate;
  • tert-butyl 3-((2-((S)-1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate;
  • (1R,3s)-3-(2-((S)-((tert-butoxycarbonyl)amino)(cyclopropyl)methyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-((tert-butoxycarbonyl)amino)(cyclopropyl)methyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-1-((tert-butoxycarbonyl)amino)ethyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(2-((S)-1-((tert-butoxycarbonyl)amino)ethyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-((tert-butoxycarbonyl)amino)(cyclopropyl)methyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-1-((tert-butoxycarbonyl)amino)ethyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(2-((S)-((tert-butoxycarbonyl)amino)(cyclopropyl)methyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(2-((S)-1-((tert-butoxycarbonyl)amino)ethyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (S)-tert-butyl (1-(5-chloro-3-(2-cyanoethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5,8-dichloro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5,8-difluoro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-cyanopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate(S)-tert-butyl ((5-chloro-3-(2-cyanoethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl ((5-chloro-3-(3-cyanopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl ((5-chloro-3-(4-cyanobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-cyanopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(4-cyanobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)carbamate;
  • (S)-tert-butyl ((5-chloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl (cyclopropyl(5,8-dichloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)methyl)carbamate;
  • (S)-tert-butyl ((8-chloro-6-fluoro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl ((5-chloro-3-(4-(methylthio)butyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(2-ethoxyethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-(dimethylamino)-3-oxopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-methoxy-3-methylbutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-morpholinopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(1,3-dimethoxypropan-2-yl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(2-(4-methylpiperazin-1-yl)ethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-4-oxo-3-(3-(piperidin-1-yl)propyl)-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • tert-butyl ((S)-1-(5-chloro-3-((S)-1-methoxypropan-2-yl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-3-(3-(4-methylpiperazin-1-yl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • (S)-tert-butyl (1-(5-chloro-4-oxo-3-(2-(pyrrolidin-1-yl)ethyl)-3,4-dihydroquinazolin-2-yl)ethyl)carbamate;
  • tert-butyl ((S)-1-(5-chloro-4-oxo-3-((S)-tetrahydrofuran-3-yl)-3,4-dihydroquinazolin-2-yl)ethyl)carbamate; and
  • (S)-tert-butyl (1-(3-(2-(1H-pyrazol-5-yl)ethyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate.

C. Preparation of a compound of formula (1) wherein n is 1, R1 is chloro, m is 0, B is (CH2)4, Ra is CONH2, R5 is hydrogen, PG is Boc, and R3 is cyclopropyl

To a mixture of (S)-tert-butyl ((5-chloro-3-(4-cyanobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate (0.42 g, 0.97 mmol) in EtOH/H2O (1:1, 10 mL) was added hydrido(dimethylphosphinous acid-kP)[hydrogen bis(dimethylphosphinito-kP)]platinum (II) (0.02 g, 0.05 mmol). The mixture stirred at 90° C. for 3 h. The mixture was cooled to room temperature and diluted with DCM/H2O. The aqueous layer was extracted with DCM. The combined organic layers were washed with brine, dried (Na2SO4), filtered and concentrated. The residue was purified via flash chromatography (SiO2, 0-20% MeOH/EtOAc) to give (S)-tert-butyl ((3-(5-amino-5-oxopentyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate. ES/MS 449.2 (M+H+).

D. Preparation of the below compound of formula (1), using the methods described in Example 1C and Reaction Scheme I:

  • tert-butyl N—[(S)-[3-(4-amino-4-oxo-butyl)-5,8-dichloro-4-oxo-quinazolin-2-yl]-cyclopropyl-methyl]carbamate; and
  • (S)-tert-butyl ((3-(4-amino-4-oxobutyl)-8-chloro-6-fluoro-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate.

E. Preparation of a compound of formula (1) wherein n is 1, R1 is chloro, m is 0, B is (CH2)2, Ra is oxiranyl, R5 is hydrogen, PG is Boc, and R3 is methyl

Solid mCPBA (1.27 g, 7.34 mmol) was added to a stirring solution of (S)-tert-butyl (1-(3-(but-3-en-1-yl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate (1.4 g, 3.67 mmol) in DCM (50 mL) at rt. The reaction mixture was stirred for 2 d then quenched with satd. NaHCO3 solution and the aqueous layer was extracted with DCM (3×10 mL). Then combined organic layers were dried over Na2SO4 and concentrated. Chromatography of the residue afforded tert-butyl ((1S)-1-(5-chloro-3-(2-(oxiran-2-yl)ethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate. ES/MS 394.1 (M+H+).

F. Preparation of a compound of formula (1) wherein n is 1, R1 is chloro, m is 0, B is hydroxy substituted butylene, Ra is NHiPr, R5 is hydrogen, PG is Boc, and R3 is methyl

Lithium perchlorate (21 mg, 0.2 mmol) and isopropylamine (1.75 mL, 20 mmol) were added to tert-butyl ((1S)-1-(5-chloro-3-(2-(oxiran-2-yl)ethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate (400 mg, 1.0 mmol). The resulting mixture was stirred for 16 h at room temperature then diluted with EtOAc (20 mL). The organic layer was washed with water, brine, dried over Na2SO4, and concentrated to give crude alcohol. Chromatography afforded tert-butyl ((1S)-1-(5-chloro-3-(3-hydroxy-4-(isopropylamino)butyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate. ES/MS 453.1 (M+H+).

G. Preparation of the below compound of Formula (1), using the methods described in Example 1F and reaction scheme I:

  • tert-butyl ((1S)-1-(3-(4-(tert-butylamino)-3-hydroxybutyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate.

H. Preparation of a compound of formula (1) wherein n is 1, R1 is chloro, m is 0, B is (CH2)4, Ra is SO2Me, R5 is hydrogen, PG is Boc, and R3 is cyclopropyl

A mixture of (S)-tert-butyl ((5-chloro-3-(4-(methylthio)butyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate (0.527 g, 1 equiv) and hydrogen peroxide (0.26 mL of 30 wt % aq, 2.2 equiv) was stirred at 75° C. for 16 h. More hydrogen peroxide (0.12 mL of 30 wt % aq, 1 equiv) was added, and the reaction mixture was stirred at 75° C. for 72 h. The mixture was cooled to rt and partitioned between DCM and H2O. The aqueous layer was extracted 2× with DCM. The combined organic layers were dried (Na2SO4), filtered, concentrated onto silica gel, and purified via flash chromatography (SiO2, 0-55% EtOAc/hexanes) to give (S)-tert-butyl ((5-chloro-3-(4-(methylsulfonyl)butyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)carbamate.

Example 2 Preparation of a Compound of Formula (2)

A. Preparation of a compound of formula (2) wherein n is 1, R1 is chloro, m is 0, and R3 is phenyl, B is (CH2)3, Ra is NHBoc, and R5 is hydrogen, and PG is Fmoc in formula (1)

To a solution of (S)-tert-butyl (3-(2-(((((9H-fluoren-9-yl)methoxy)carbonyl)amino)(phenyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate (0.28 g, 0.42 mmol) in DMF (2 mL) was added piperidine (0.15 mL, 1.5 mmol). The mixture stirred at room temperature for 2 hours. The mixture was diluted with EtOAc/H2O and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with brine, dried (Na2SO4), decanted and concentrated. The residue was purified via flash chromatography (SiO2, 0-20% MeOH/DCM) to give tert-butyl N-[3-[2-[(S)-amino(phenyl)methyl]-5-chloro-4-oxo-quinazolin-3-yl]propyl]carbamate. ES/MS 443.2 (M+H+).

B. Preparation of the below compound of formula (2), using the methods described in Example 2A and Reaction Scheme I and PG is Fmoc in formula (1)

  • (S)-tert-butyl (4-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)-2-methylbutan-2-yl)carbamate;
  • (S)-tert-butyl (2-((2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)(benzyl)amino)ethyl)carbamate;
  • tert-butyl (2-(2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (2-((2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)(methyl)amino)ethyl)carbamate;
  • tert-butyl (3-(2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl)carbamate;
  • (S)-tert-butyl (2-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)ethyl)carbamate;
  • tert-butyl (3-(2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)-2-methylpropyl)carbamate;
  • (S)-tert-butyl (4-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl)carbamate
  • (S)-tert-butyl (3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)(methyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-amino-3-methylbutyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate
  • tert-butyl N-[3-[2-[(1S)-1-aminopropyl]-5-chloro-4-oxo-quinazolin-3-yl]propyl]carbamate;
  • tert-butyl N-[3-[2-[(S)-amino(cyclopropyl)methyl]-5-chloro-4-oxo-quinazolin-3-yl]propyl]carbamate;
  • tert-butyl N-[3-[2-[(1S)-1-aminoethyl]-5-chloro-4-oxo-quinazolin-3-yl]propyl]-N-methyl-carbamate;
  • tert-butyl N-[3-[2-[(1S)-1-aminoethyl]-5-chloro-4-oxo-quinazolin-3-yl]propyl]carbamate;
  • tert-butyl N-[3-[2-[(S)-amino(cyclopropyl)methyl]-5-chloro-4-oxo-quinazolin-3-yl]propyl]-N-methyl-carbamate;
  • (S)-tert-butyl (3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl)carbamate;
  • (S)-tert-butyl 4-((2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)morpholine-4-carboxylate;
  • tert-butyl 3-((2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)pyrrolidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)pyrrolidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate; and
  • tert-butyl 3-((2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate.

C. Preparation of a compound of formula (2) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, Ra is —N(CH3)2, R5 is hydrogen, and R3 is methyl and PG is Boc in formula (1)

Trifluoroacetic acid (2 mL) was added to a solution of (S)-tert-butyl (1-(5-chloro-3-(3-(dimethylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate (0.5 g, 1.6 mmol) in dichloromethane (3 mL). The reaction mixture was stirred at room temperature for 1 hour. The solvents was removed in vacuo to afford (S)-2-(1-aminoethyl)-5-chloro-3-(3-(dimethylamino)propyl)quinazolin-4(3H)-one 2,2,2-trifluoroacetic acid salt. ES/MS m/z=309.2 (M+H)+

Preparation of the below compounds of formula (2), using the methods described in Example 2c and Reaction Scheme I and PG is Boc in formula (1):

  • (S)-2-(1-aminoethyl)-5-chloro-3-(3-morpholinopropyl)quinazolin-4(3H)-one;
  • 2-((S)-1-aminoethyl)-5-chloro-3-(3-hydroxy-4-(isopropylamino)butyl)quinazolin-4(3H)-one;
  • 2-((S)-1-aminoethyl)-3-(4-(tert-butylamino)-3-hydroxybutyl)-5-chloroquinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(2-(dimethylamino)ethyl)quinazolin-4(3H)-one;
  • (S)-3-(2-(amino(cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propanenitrile;
  • (S)-5-(2-(amino(cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)pentanamide;
  • 2-[(1S)-1-aminoethyl]-3-[3-[benzyl(methyl)amino]propyl]-5-chloro-quinazolin-4-one;
  • 4-[2-[(S)-amino(cyclopropyl)methyl]-5-chloro-4-oxo-quinazolin-3-yl]butanenitrile;
  • 5-[2-[(S)-amino(cyclopropyl)methyl]-5-chloro-4-oxo-quinazolin-3-yl]pentanamide;
  • 4-[2-[(S)-amino(cyclopropyl)methyl]-5,8-dichloro-4-oxo-quinazolin-3-yl]butanamide;
  • 2-[3-[2-[(S)-amino(cyclopropyl)methyl]-5-chloro-4-oxo-quinazolin-3-yl]phenyl]acetonitrile;
  • (S)-3-((1H-pyrazol-3-yl)methyl)-2-(1-aminoethyl)-5-chloroquinazolin-4(3H)-one;
  • (S)-3-((1H-pyrazol-4-yl)methyl)-2-(1-aminoethyl)-5-chloroquinazolin-4(3H)-one;
  • (S)-3-((1H-imidazol-2-yl)methyl)-2-(1-aminoethyl)-5-chloroquinazolin-4(3H)-one;
  • (S)-3-((1H-1,2,4-triazol-5-yl)methyl)-2-(1-aminoethyl)-5-chloroquinazolin-4(3H)-one;
  • (1R,3s)-3-(2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (S)-3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl benzoate;
  • 3-(2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl benzoate;
  • (1S,4r)-4-(2-((S)-1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclohexyl benzoate;
  • (1R,3s)-3-(2-((S)-amino(cyclopropyl)methyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-amino(cyclopropyl)methyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-1-aminoethyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(2-((S)-1-aminoethyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-amino(cyclopropyl)methyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(2-((S)-1-aminoethyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(2-((S)-amino(cyclopropyl)methyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(2-((S)-1-aminoethyl)-5,8-dichloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (S)-3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propanenitrile;
  • (S)-3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propanenitrile;
  • (S)-2-(1-aminoethyl)-5,8-dichloro-3-(3-methoxypropyl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5,8-difluoro-3-(3-methoxypropyl)quinazolin-4(3H)-one;
  • (S)-4-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butanenitrile;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(3-(methylsulfonyl)propyl)quinazolin-4(3H)-one;
  • (S)-3-(2-(amino(cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propanenitrile(S)-4-(2-(amino(cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butanenitrile;
  • (S)-5-(2-(amino(cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)pentanenitrile;
  • (S)-4-(2-(1-aminopropyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butanenitrile;
  • (S)-5-(2-(1-aminopropyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)pentanenitrile;
  • (S)-2-(1-aminopropyl)-5-chloro-3-(3-(methylsulfonyl)propyl)quinazolin-4(3H)-one;
  • (S)-2-(amino(cyclopropyl)methyl)-5-chloro-3-(3-(methylsulfonyl)propyl)quinazolin-4(3H)-one;
  • (S)-2-(amino(cyclopropyl)methyl)-5,8-dichloro-3-(3-(methylsulfonyl)propyl)quinazolin-4(3H)-one;
  • (S)-2-(amino(cyclopropyl)methyl)-8-chloro-6-fluoro-3-(3-(methylsulfonyl)propyl)quinazolin-4(3H)-one;
  • (S)-4-(2-(amino(cyclopropyl)methyl)-8-chloro-6-fluoro-4-oxoquinazolin-3(4H)-yl)butanamide;
  • (S)-2-(amino(cyclopropyl)methyl)-5-chloro-3-(4-(methylthio)butyl)quinazolin-4(3H)-one;
  • (S)-3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)-N,N-dimethylpropanamide;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(2-ethoxyethyl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(3-methoxy-3-methylbutyl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(3-morpholinopropyl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(1,3-dimethoxypropan-2-yl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(2-(4-methylpiperazin-1-yl)ethyl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(3-(piperidin-1-yl)propyl)quinazolin-4(3H)-one;
  • 2-((S)-1-aminoethyl)-5-chloro-3-((S)-1-methoxypropan-2-yl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(2-(pyrrolidin-1-yl)ethyl)quinazolin-4(3H)-one;
  • (S)-2-(1-aminoethyl)-5-chloro-3-(3-(4-methylpiperazin-1-yl)propyl)quinazolin-4(3H)-one;
  • 2-((S)-1-aminoethyl)-5-chloro-3-((S)-tetrahydrofuran-3-yl)quinazolin-4(3H)-one; and
  • (S)-3-(2-(1H-pyrazol-5-yl)ethyl)-2-(1-aminoethyl)-5-chloroquinazolin-4(3H)-one.

Example 3 Preparation of Compounds of Formula (I) or (3)

A. Preparation of a compound of formula (3) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, Ra is NHBoc, R5 is hydrogen, A is a single bond, R4 is 2,6-diamino-5-cyanopyrimidine, and R3 is phenyl

A mixture of (S)-tert-butyl (3-(2-(amino(phenyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate (71 mg, 0.16 mmol), 2,4-diamino-6-chloropyrimidine-5-carbonitrile (26 mg, 0.16 mmol), and diisopropylethylamine (42 μL, 0.24 mmol) in i-PrOH (1 mL) was heated in the microwave at 130° C. for 8 h. On cooling, the mixture was concentrated onto silica gel and purified via flash chromatography (SiO2, 0-100% EtOAc/hexanes, then 0-20% MeOH/EtOAc) to give (S)-tert-butyl (3-(5-chloro-2-(((2,6-diamino-5-cyanopyrimidin-4-yl)amino)(phenyl)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate. ES/MS 576.2 (M+H+).

B. Preparation of the below compounds of formulae (I) or (3), using the methods described in Example 3A and Reaction Scheme I:

  • (S)-tert-butyl (4-(5-chloro-4-oxo-2-(1-(thiazolo[5,4-d]pyrimidin-7-ylamino)ethyl)quinazolin-3(4H)-yl)-2-methylbutan-2-yl)carbamate;
  • (S)-tert-butyl (4-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)-2-methylbutan-2-yl)carbamate;
  • (S)-tert-butyl (3-(5-chloro-2-(1-((2,6-diamino-5-chloropyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (2-(benzyl(5-chloro-2-(1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)amino)ethyl)carbamate;
  • (S)-tert-butyl (2-((5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)-4-oxoquinazolin-3(4H)-yl)(methyl)amino)ethyl)carbamate;
  • (S)-tert-butyl (2-((2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)(benzyl)amino)ethyl)carbamate;
  • (S)-tert-butyl (2-((5-chloro-2-(1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)(methyl)amino)ethyl)carbamate;
  • (S)-tert-butyl (2-((2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)(methyl)amino)ethyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-(3-aminopyrazine-2-carboxamido)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-methyl-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((6-amino-5-chloropyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)(methyl)carbamate
  • tert-butyl (3-(2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl)carbamate;
  • tert-butyl (2-(2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (2-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)ethyl)carbamate;
  • tert-butyl (3-(2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)-2-methylpropyl)carbamate;
  • (S)-tert-butyl (4-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)-3-methylbutyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-aminopropyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • tert-butyl N-[3-[2-[(1S)-1-[(6-amino-5-cyano-pyrimidin-4-yl)amino]propyl]-5-chloro-4-oxo-quinazolin-3-yl]propyl]carbamate;
  • tert-butyl N-[3-[5-chloro-2-[(S)-cyclopropyl-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)methyl]-4-oxo-quinazolin-3-yl]propyl]carbamate;
  • (S)-tert-butyl (3-(5-chloro-2-(1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)propyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(5-chloro-2-(cyclopropyl(pyrido[3,2-d]pyrimidin-4-ylamino)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(5-chloro-2-(cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • tert-butyl N-[3-[5-chloro-2-[(1S)-1-[(2,6-diamino-5-cyano-pyrimidin-4-yl)amino]ethyl]-4-oxo-quinazolin-3-yl]propyl]-N-methyl-carbamate;
  • (S)-tert-butyl (3-(2-(((6-amino-5-cyanopyrimidin-4-yl)amino)(cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((2-amino-5-cyano-6-methylpyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)(methyl)carbamate;
  • tert-butyl N-[3-[5-chloro-2-[(1S)-1-[(2,6-diamino-5-cyano-pyrimidin-4-yl)amino]ethyl]-4-oxo-quinazolin-3-yl]propyl]carbamate;
  • (S)-2-amino-4-chloro-6-(((5-chloro-3-(3-(methylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl) (cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile;
  • (S)-tert-butyl (3-(5-chloro-2-(cyclopropyl(thiazolo[5,4-d]pyrimidin-7-ylamino)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(((5-bromo-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) (cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • tert-butyl N-[3-[5-chloro-2-[(S)-cyclopropyl-[(2,6-diamino-5-cyano-pyrimidin-4-yl)amino]methyl]-4-oxo-quinazolin-3-yl]propyl]-N-methyl-carbamate;
  • (S)-tert-butyl (3-(5-chloro-2-(cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(5-chloro-2-(((6-chloropyrido[3,2-d]pyrimidin-4-yl)amino) (cyclopropyl)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(((2-amino-5-cyano-6-methylpyrimidin-4-yl)amino) (cyclopropyl)methyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(5-chloro-4-oxo-2-(1-(thiazolo[5,4-d]pyrimidin-7-ylamino)ethyl)quinazolin-3(4H)-yl)propyl)carbamate;
  • (S)-tert-butyl (3-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate;
  • (1R,3s)-3-(2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (S)-3-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl benzoate;
  • 3-(2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)butyl benzoate;
  • (1S,4r)-4-(2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclohexyl benzoate;
  • (S)-tert-butyl (3-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl)carbamate;
  • (S)-tert-butyl 4-((2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)morpholine-4-carboxylate;
  • tert-butyl 3-((2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)pyrrolidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)pyrrolidine-1-carboxylate;
  • tert-butyl 2-((2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate;
  • tert-butyl 3-((2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate;
  • (1R,3s)-3-(5-chloro-2-((S)-1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(5-chloro-2-((S)-1-((2,6-diamino-5-chloropyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(5,8-dichloro-2-((S)-cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(5,8-dichloro-2-((S)-cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(8-chloro-2-((S)-cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(8-chloro-2-((S)-1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(8-chloro-2-((S)-1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(5,8-dichloro-2-((S)-cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1S,3r)-3-(5,8-dichloro-2-((S)-1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(8-chloro-2-((S)-cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-6-fluoro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (1R,3s)-3-(5,8-dichloro-2-((S)-1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate;
  • (R)-tert-butyl 3-((5-chloro-2-((S)-1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)methyl)pyrrolidine-1-carboxylate; and
  • (S)-tert-butyl 3-((5-chloro-2-((S)-1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)methyl)piperidine-1-carboxylate.

C. Preparation of a compound of formula (3), where A is CO, n is 1, R1 is Cl, m is 0, B is (CH2)3, Ra is NHBoc, R5 is hydrogen, R4 is 2,4-diamino-5-bromopyrazine, and R3 is methyl

To (S)-tert-butyl (3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate and 3,5-diamino-6-bromopyrazine-2-carboxylic acid in DCM was added HATU and DIEA. The reaction was allowed to stir at RT for 2 hours. The reaction was monitored by LCMS. Upon completion the reaction was concentrated to dryness and purified by silica gel chromatography (20-80% EtOAc/hexanes) to give (S)-tert-butyl (3-(5-chloro-2-(1-(3,5-diamino-6-bromopyrazine-2-carboxamido)ethyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate.

D. Preparation of the below compound of formula (3), where A is CO, n is 1, R1 is Cl, m is 0, B is (CH2)3, Ra is NHBoc, R5 is hydrogen, R4 is 2-aminopyrazolo[1,5-a]pyrimidine-3-yl, and R3 is methyl

To (S)-tert-butyl (3-(2-(1-aminoethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate (300 mg, 0.79 mmol), HATU (400 mg, 1.1 mmol) and 2-aminopyrazolo[1,5-a]pyrimidine-3-carboxylic acid (196 mg, 1.1 mmol) in DMF (3 mL) was added Hunig's base (0.4 mL). After stirring at room temp for 1 hour, water was added and the mixture was extracted with EtOAc (3×30 mL), dried (MgSO4), and concentrated. The residue was purified on a reverse phase HPLC run from 0 to 95% ACN in water (0.1% TFA) to give (S)-tert-butyl (3-(2-(1-(2-aminopyrazolo[1,5-a]pyrimidine-3-carboxamido)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)carbamate. ES/MS 541.2 (M+H)+.

E. Preparation of a compound of formula (I), where n is 1, R1 is chloro, m is 0, B is (CH2)3, R2 is NH2, R5 is hydrogen, R4 is 2,6-diamino-5-cyanopyrimidin-4-yl, R3 is phenyl, and R3 is phenyl

To a solution of (S)-tert-butyl (3-(5-chloro-2-(((2,6-diamino-5-cyanopyrimidin-4-yl)amino)(phenyl)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)carbamate (53 mg, 0.09 mmol) in DCM (0.5 mL) was added trifluoroacetic acid (0.5 mL). The mixture sat at room temperature 2 h. The mixture was concentrated to give (S)-2,4-diamino-6-(((3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)(phenyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 1). 1H NMR (400 MHz, DMSO-d6) δ 7.80 (t, J=8.0 Hz, 1H), 7.76-7.67 (m, 3H), 7.61 (dd, J=8.0, 1.2 Hz, 1H), 7.60 (dd, J=8.1, 1.2 Hz, 1H), 7.53-7.48 (m, 2H), 7.43-7.34 (m, 3H), 7.10 (br s, 2H), 6.84 (br s, 2H), 6.49 (d, J=7.4 Hz, 1H), 4.18-4.03 (m, 1H), 3.85-3.76 (m, 1H), 2.91-2.78 (m, 2H), 2.07-1.94 (m, 1H), 1.79-1.62 (m, 1H). ES/MS 476.2 (M+H+).

F. Preparation of the below compounds of formula (I), using Example 3E and Reaction Scheme I:

  • (S)-3-(3-amino-3-methylbutyl)-5-chloro-2-(1-(thiazolo[5,4-d]pyrimidin-7-ylamino)ethyl)quinazolin-4(3H)-one (Compound 2). 1H NMR (400 MHz, DMSO-d6) δ 9.30 (s, 1H), 8.81 (d, J=6.8 Hz, 1H), 8.43 (s, 1H), 7.90 (br s, 3H), 7.68 (dd, J=8.4, 7.6 Hz, 1H), 7.57-7.46 (m, 2H), 5.50 (p, J=6.8 Hz, 1H), 4.21 (td, J=13.4, 4.2 Hz, 2H), 4.00 (td, J=12.2, 4.7 Hz, 2H), 2.14 (td, J=13.2, 4.8 Hz, 2H), 1.98 (td, J=12.6, 4.0 Hz, 2H), 1.63 (d, J=6.5 Hz, 3H), 1.24 (s, 3H), 1.23 (s, 3H). ES/MS 444.1 (M+H+).
  • (S)-4-amino-6-((1-(3-(3-amino-3-methylbutyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 3). 1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1H), 7.93-7.84 (m, 2H), 7.71 (t, J=8.1 Hz, 1H), 7.60-7.50 (m, 2H), 5.40 (p, J=6.6 Hz, 1H), 4.09 (td, J=13.3, 4.8 Hz, 2H), 3.93 (td, J=13.3, 4.8 Hz, 2H), 2.04 (td, J=12.6, 4.7 Hz, 2H), 1.94 (td, J=12.5, 4.5 Hz, 2H), 1.52 (d, J=6.6 Hz, 3H), 1.26 (s, 3H), 1.25 (s, 3H). ES/MS 427.2 (M+H+).
  • (S)-4-amino-6-((1-(3-(3-(benzylamino)propyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 6). 1H NMR (400 MHz, DMSO-d6) δ 8.78 (br s, 2H), 7.97 (s, 1H), 7.78 (d, J=6.8 Hz, 1H), 7.70 (dd, J=8.4, 7.6 Hz, 1H), 7.55-7.35 (m, 8H), 5.34 (p, J=6.8 Hz, 1H), 4.23-4.09 (m, 3H), 3.99 (p, J=7.1 Hz, 1H), 3.03 (p, J=7.0 Hz, 2H), 2.26-2.02 (m, 2H), 1.51 (d, J=6.6 Hz, 3H). ES/MS 489.2 (M+H+).
  • (S)-3-(3-aminopropyl)-5-chloro-2-(1-((2,6-diamino-5-chloropyrimidin-4-yl)amino)ethyl)quinazolin-4(3H)-one (Compound 7). 1H NMR (400 MHz, DMSO-d6) δ 7.79-7.68 (m, 3H), 7.58 (ddd, J=7.6, 6.6, 1.2 Hz, 2H), 5.35 (p, J=7.5 Hz, 1H), 4.18-4.00 (m, 2H), 2.98-2.86 (m, 2H), 2.07-1.93 (m, 2H), 1.56 (d, J=6.5 Hz, 3H). ES/MS 423.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-(3-(benzylamino)propyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 8). 1H NMR (400 MHz, DMSO-d6) δ 8.89-8.73 (m, 2H), 7.75 (dd, J=8.3, 7.8 Hz, 1H), 7.58 (ddd, J=9.9, 8.1, 1.2 Hz, 2H), 7.52-7.39 (m, 5H), 7.21-7.10 (m, 2H), 6.95-6.79 (m, 2H), 5.40 (p, J=6.8 Hz, 1H), 4.19-4.02 (m, 4H), 3.07 (s, 2H), 2.21-2.02 (m, 2H), 1.53 (d, J=6.6 Hz, 3H). ES/MS 504.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-((2-aminoethyl)(benzyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 9). 1H NMR (400 MHz, DMSO-d6) δ 7.78 (t, J=8.0 Hz, 1H), 7.72 (s, 4H), 7.61 (dd, J=7.9, 1.1 Hz, 1H), 7.56 (dd, J=8.1, 1.2 Hz, 1H), 7.30 (dd, J=6.6, 2.9 Hz, 2H), 7.20-7.16 (m, 3H), 5.72 (p, J=6.7 Hz, 1H), 4.63 (d, J=12.9 Hz, 1H), 4.49 (d, J=12.8 Hz, 1H), 3.70-3.53 (m, 2H), 2.89 (dt, J=11.7, 6.3 Hz, 2H), 1.46 (d, J=6.6 Hz, 3H). ES/MS 505.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-((2-aminoethyl)(benzyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 10). 1H NMR (400 MHz, DMSO-d6) δ 7.78 (t, J=8.0 Hz, 1H), 7.63-7.56 (m, 2H), 7.45-7.39 (m, 4H), 7.38-7.28 (m, 1H), 5.88 (p, J=7.2 Hz, 1H), 4.77 (d, J=13.7 Hz, 1H), 4.50 (d, J=13.7 Hz, 1H), 3.70 (td, J=11.6, 10.9, 5.4 Hz, 1H), 3.32 (td, J=12.4, 11.9, 5.3 Hz, 1H), 2.95 (dd, J=13.3, 7.6 Hz, 2H), 1.25 (d, J=6.6 Hz, 3H). ES/MS 505.2 (M+H+).
  • (S)-3-((2-aminoethyl)(methyl)amino)-5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)quinazolin-4(3H)-one (Compound 11). Exists as a ˜1:1 ratio of diastereomers. 1H NMR (400 MHz, DMSO-d6) δ 9.26 (d, J=7.3 Hz, 0.5H), 9.19 (d, J=6.8 Hz, 0.5H), 8.13 (d, J=4.4 Hz, 1H), 8.10 (d, J=1.1 Hz, 1H), 7.88 (s, 1.5H), 7.81 (s, 2H), 7.74-7.63 (m, 2H), 7.56 (q, J=1.2 Hz, 0.5H), 7.54 (dd, J=1.7, 1.0 Hz, 0.5H), 7.53 (dd, J=7.9, 1.2 Hz, 0.5H), 7.48 (dd, J=8.2, 1.2 Hz, 0.5H), 5.89-5.81 (m, 1H), 3.83 (dt, J=12.3, 5.9 Hz, 0.5H), 3.68 (ddd, J=12.9, 9.3, 6.4 Hz, 0.5H), 3.63-3.51 (m, 0.5H), 3.37-3.20 (m, 1H), 3.16-3.00 (m, 3.5H), 2.95 (s, 1.5H), 1.62 (d, J=6.9 Hz, 1.5H), 1.60 (d, J=6.9 Hz, 1.5H). ES/MS 414.1 (M+H+).
  • (S)-4-amino-6-((1-(3-((2-aminoethyl)(benzyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 12). 1H NMR (400 MHz, DMSO-d6) δ 8.05 (s, 1H), 7.70 (t, J=8.0 Hz, 1H), 7.64-7.56 (m, 3H), 7.54 (dd, J=7.9, 1.2 Hz, 1H), 7.51 (dd, J=8.1, 1.2 Hz, 1H), 7.45-7.27 (m, 8H), 5.84 (p, J=6.7 Hz, 1H), 4.79 (d, J=14.0 Hz, 1H), 4.47 (d, J=14.0 Hz, 1H), 3.70 (td, J=13.1, 10.9, 5.8 Hz, 1H), 3.30 (td, J=12.9, 10.6, 5.6 Hz, 1H), 3.09-2.94 (m, 2H), 1.27 (d, J=6.8 Hz, 3H). ES/MS 490.2 (M+H+).
  • (S)-4-amino-6-((1-(3-((2-aminoethyl)(benzyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 13). 1H NMR (400 MHz, DMSO-d6) δ 8.04 (d, J=0.5 Hz, 1H), 7.74 (t, J=8.0 Hz, 1H), 7.68-7.63 (m, 4H), 7.57 (dd, J=7.9, 1.1 Hz, 1H), 7.53 (dd, J=8.1, 1.1 Hz, 1H), 7.34 (s, 2H), 7.27 (dd, J=7.5, 1.9 Hz, 2H), 7.17-7.07 (m, 3H), 5.76 (p, J=6.6 Hz, 1H), 4.61 (d, J=12.6 Hz, 1H), 4.49 (d, J=12.5 Hz, 1H), 3.67-3.48 (m, 2H), 2.92-2.72 (m, 2H), 1.45 (d, J=6.6 Hz, 3H). ES/MS 490.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-((2-aminoethyl)(methyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 14). 1H NMR (400 MHz, DMSO-d6) δ 7.80-7.71 (m, 3H), 7.60 (dd, J=8.2, 1.2 Hz, 1H), 7.57 (dd, J=7.8, 1.2 Hz, 1H), 5.62 (p, J=7.2 Hz, 1H), 3.74 (dt, J=12.6, 6.2 Hz, 1H), 3.30 (dt, J=13.2, 8.2 Hz, 1H), 3.05-2.96 (m, 2H), 2.91 (s, 3H), 1.46 (d, J=6.7 Hz, 3H). ES/MS 429.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-((2-aminoethyl)(methyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 15). 1H NMR (400 MHz, DMSO-d6) δ 7.79-7.71 (m, 4H), 7.56 (ddd, J=8.1, 4.0, 1.2 Hz, 2H), 5.64 (p, J=6.8 Hz, 1H), 3.65-3.55 (m, 1H), 3.52-3.44 (m, 1H), 3.07 (s, 4H), 3.00-2.90 (m, 1H), 1.48 (d, J=6.7 Hz, 3H). ES/MS 429.1 (M+H+).
  • (S)-4-amino-6-((1-(3-((2-aminoethyl)(methyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 16). 1H NMR (400 MHz, DMSO-d6) δ 8.01 (s, 1H), 7.84 (br s, 3H), 7.75-7.69 (m, 2H), 7.54 (ddd, J=13.6, 8.0, 1.2 Hz, 2H), 5.68 (p, J=6.8 Hz, 1H), 3.74 (dt, J=12.0, 5.7 Hz, 1H), 3.24 (dt, J=13.5, 8.2 Hz, 1H), 3.04-2.94 (m, 2H), 2.86 (s, 3H), 1.44 (d, J=6.7 Hz, 3H). ES/MS 414.1 (M+H+).
  • (S)-4-amino-6-((1-(3-((2-aminoethyl)(methyl)amino)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 17). 1H NMR (400 MHz, DMSO-d6) δ 8.00 (s, 1H), 7.72 (s, 4H), 7.68 (t, J=8.0 Hz, 1H), 7.56 (d, J=7.1 Hz, 1H), 7.49 (ddd, J=8.1, 6.0, 1.2 Hz, 2H), 7.36 (s, 2H), 5.67 (p, J=7.0 Hz, 1H), 3.62-3.52 (m, 1H), 3.52-3.43 (m, 1H), 3.14 (t, J=13.1 Hz, 1H), 3.06-2.95 (m, 4H), 1.47 (d, J=6.8 Hz, 3H). ES/MS 414.1 (M+H+).
  • (S)-3-amino-N-(1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)pyrazine-2-carboxamide (Compound 18). 1H NMR (400 MHz, DMSO-d6) δ 9.08 (d, J=7.3 Hz, 1H), 8.23 (d, J=2.3 Hz, 1H), 7.86 (d, J=2.3 Hz, 1H), 7.73-7.67 (m, 4H), 7.57-7.49 (m, 2H), 5.24 (p, J=6.7 Hz, 1H), 4.25 (ddd, J=14.2, 8.5, 5.9 Hz, 1H), 4.00 (ddd, J=14.7, 8.9, 6.5 Hz, 1H), 2.92 (h, J=7.0 Hz, 2H), 2.12-1.92 (m, 2H), 1.53 (d, J=6.6 Hz, 3H). ES/MS 402.1 (M+H+).
  • (S)-4-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-6-methylpyrimidine-5-carbonitrile (Compound 23). 1H NMR (400 MHz, DMSO-d6) δ 8.52 (s, 1H), 7.73 (d, J=8.0 Hz, 3H), 7.55 (d, J=4.7 Hz, 1H), 5.40 (m, 1H), 4.23 (s, 1H), 4.00 (s, 1H), 3.46 (brm, 2H), 2.93 (m, 2H), 2.48 (s, 3H), 2.24-2.11 (m, 1H), 2.07 (m, 1H), 1.58 (d, J=6.6 Hz, 3H). ES/MS 398.1 (M+H+).
  • 4-amino-6-(((1S)-1-(3-(4-aminobutan-2-yl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 24). 1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1H), 7.76 (d, J=7.1 Hz, 1H), 7.69-7.61 (m, 1H), 7.50-7.43 (m, 2H), 7.35 (s, 1H), 5.46 (t, J=6.8, 6.8 Hz, 1H), 4.54 (m, 1H), 2.95 (m, 1H), 2.66 (m, 1H), 2.55 (m, 1H), 2.16-2.04 (m, 1H), 1.57 (d, J=6.5 Hz, 3H), 1.47 (d, J=6.7 Hz, 3H). ES/MS 413.0 (M+H+).
  • 4-amino-6-(((1S)-1-(3-(1-aminopropan-2-yl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 25). 1H NMR (400 MHz, DMSO-d6) δ 8.08 (s, 1H), 7.71 (t, J=8.0, 8.0 Hz, 1H), 7.57-7.49 (m, 2H), 7.35 (s, 2H), 5.73 (m, 1H), 4.33 (m, 1H), 3.69 (m, 1H), 3.23 (m, 1H), 1.50 (d, J=6.2 Hz, 3H), 1.45 (d, J=6.4 Hz, 3H). ES/MS 398.9 (M+H+).
  • (S)-4-amino-6-((1-(3-(2-aminoethyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 26). 1H NMR (400 MHz, DMSO-d6) δ 7.95 (s, 1H), 7.80 (s, 2H), 7.76 (d, J=6.6 Hz, 1H), 7.70 (t, J=8.0, 8.0 Hz, 1H), 7.54-7.49 (m, 2H), 7.37 (s, 1H), 5.29 (m, 1H), 4.59-4.46 (m, 1H), 4.07 (m, 1H), 1.51 (d, J=6.7 Hz, 3H). ES/MS 385.0 (M+H+).
  • 4-amino-6-(((1S)-1-(3-(3-amino-2-methylpropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 27). 1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1H), 7.73-7.68 (m, 2H), 7.63 (m, 1H), 7.56-7.51 (m, 2H), 5.36 (m, 1H), 4.22 (m, 1H), 4.06-3.95 (m, 2H), 3.01-2.92 (m, 1H), 2.73 (m, 1H), 1.51 (d, J=6.6 Hz, 3H), 1.00 (dd, J=6.8, 2.5 Hz, 3H). ES/MS 413.0 (M+H+).
  • (S)-4-amino-6-((1-(3-(4-aminobutyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 28). 1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1H), 7.75 (m, 1H), 7.66 (m, 1H), 7.61 (s, 2H), 7.56-7.47 (m, 2H), 7.36 (s, 2H), 5.42 (m, 1H), 4.14-4.00 (m, 1H), 3.95-3.81 (m, 1H), 2.78 (m, 2H), 1.76 (m, 2H), 1.56 (m, 2H), 1.51 (d, J=6.6 Hz, 3H). ES/MS 413.0 (M+H+).
  • (S)-4-amino-6-((1-(3-(3-aminopropyl)-5-methyl-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 29). 1H NMR (400 MHz, DMSO-d6) δ 8.15 (s, 1H), 7.77 (d, J=7.2 Hz, 1H), 7.68-7.57 (m, 1H), 7.49 (s, 3H), 7.41 (d, J=8.1 Hz, 1H), 7.26 (dd, J=7.6, 1.3 Hz, 1H), 5.42 (p, J=6.7 Hz, 1H), 4.23 (ddd, J=14.4, 8.8, 5.8 Hz, 1H), 4.01 (ddd, J=14.5, 8.7, 6.1 Hz, 1H), 2.91 (dd, J=10.5, 4.4 Hz, 2H), 2.75 (s, 3H), 2.11 (ddq, J=43.7, 14.1, 6.6 Hz, 2H), 1.56 (d, J=6.6 Hz, 3H). ES/MS 379.2 (M+H+).
  • (S)-2-(1-((6-amino-5-chloropyrimidin-4-yl)amino)ethyl)-3-(3-aminopropyl)-5-chloroquinazolin-4(3H)-one (Compound 30). 1H NMR (400 MHz, DMSO-d6) δ 7.93 (s, 1H), 7.85-7.63 (m, 3H), 7.54 (ddt, J=8.4, 1.8, 0.9 Hz, 1H), 7.11 (d, J=7.1 Hz, 1H), 6.89 (s, 2H), 5.36 (p, J=6.7 Hz, 1H), 4.26 (ddd, J=14.2, 8.5, 5.9 Hz, 1H), 4.02 (ddd, J=14.5, 8.9, 6.1 Hz, 1H), 2.93 (h, J=7.0 Hz, 2H), 2.17 (dt, J=14.7, 7.3 Hz, 1H), 2.06 (dq, J=13.9, 7.0, 6.6 Hz, 1H), 1.55 (d, J=6.6 Hz, 2H). ES/MS 408.1 (M+H+).
  • (S)-4-amino-6-((1-(5-chloro-3-(3-(methylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 31) 1H NMR (400 MHz, DMSO-d6) δ 8.18 (s, 2H), 7.98 (s, 3H), 7.73 (dd, J=8.5, 7.5 Hz, 3H), 7.65 (s, 3H), 7.59-7.51 (m, 2H), 5.42 (p, J=6.7 Hz, 1H), 4.23 (ddd, J=14.4, 8.8, 5.8 Hz, 1H), 4.01 (ddd, J=14.5, 8.7, 6.1 Hz, 1H), 2.91 (dd, J=10.5, 4.4 Hz, 2H), 2.61 (t, J=5.1 Hz, 3H), 2.11 (ddq, J=43.7, 14.1, 6.6 Hz, 2H), 1.56 (d, J=6.6 Hz, 3H). ES/MS 413.2 (M+H+).
  • (S)-2-amino-N-(1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (Compound 32) 1H NMR (400 MHz, DMSO-d6) δ 8.91 (dd, J=6.7, 1.6 Hz, 1H), 8.64-8.57 (m, 2H), 7.75-7.66 (m, 3H), 7.55 (ddd, J=22.8, 8.0, 1.2 Hz, 2H), 7.01 (dd, J=6.7, 4.5 Hz, 1H), 6.49 (s, 2H), 5.38 (p, J=6.7 Hz, 1H), 4.33 (ddd, J=14.5, 8.6, 5.8 Hz, 1H), 4.09-3.97 (m, 1H), 3.00-2.87 (m, 2H), 2.02 (dh, J=28.2, 6.8 Hz, 2H), 1.53 (d, J=6.5 Hz, 3H). ES/MS 441.2 (M+H+).
  • (S)-4-amino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-3-methylbutyl)amino)pyrimidine-5-carbonitrile (Compound 33). 1H NMR (400 MHz, DMSO-d6) δ 8.04 (s, 1H), 7.79-7.69 (m, 4H), 7.54 (d, J=8.2 Hz, 1H), 7.41 (br s, 2H), 5.36 (ddd, J=10.9, 7.2, 2.8 Hz, 1H), 4.41-4.24 (m, 1H), 3.94-3.77 (m, 1H), 3.00-2.83 (m, 2H), 2.26-1.92 (m, 3H), 1.89-1.74 (m, 1H), 1.64 (ddd, J=13.2, 9.7, 3.1 Hz, 1H), 0.98 (d, J=6.5 Hz, 3H), 0.94 (d, J=6.6 Hz, 3H). ES/MS 441.2 (M+H+).
  • (S)-4-amino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)amino)pyrimidine-5-carbonitrile (Compound 34). 1H NMR (400 MHz, DMSO-d6) δ 8.06 (s, 1H), 7.78-7.70 (m, 3H), 7.59-7.52 (m, 2H), 7.46 (br s, 2H), 5.26 (q, J=7.1 Hz, 1H), 4.31-4.16 (m, 1H), 4.08-3.93 (m, 1H), 3.03-2.86 (m, 2H), 2.24-1.88 (m, 4H), 1.00 (t, J=7.3 Hz, 3H). ES/MS 413.1 (M+H+). (S)-3-(3-aminopropyl)-5-chloro-2-(cyclopropyl(imidazo[2,1-][1,2,4]triazin-4-ylamino)methyl)quinazolin-4(3H)-one (Compound 35). 1H NMR (400 MHz, DMSO-d6) δ 9.16 (d, J=7.1 Hz, 1H), 8.15 (s, 1H), 8.13 (d, J=1.1 Hz, 1H), 7.74 (dd, J=8.2, 7.7 Hz, 1H), 7.70 (s, 2H), 7.67 (d, J=1.1 Hz, 1H), 7.59 (dd, J=8.2, 1.2 Hz, 1H), 7.57 (dd, J=7.8, 1.2 Hz, 1H), 5.13 (t, J=7.9 Hz, 1H), 4.28-4.05 (m, 2H), 2.99-2.85 (m, 2H), 2.22-1.94 (m, 2H), 1.81-1.67 (m, 1H), 0.68-0.57 (m, 3H), 0.56-0.46 (m, 1H). ES/MS 425.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)amino)pyrimidine-5-carbonitrile (Compound 36). 1H NMR (400 MHz, DMSO-d6) δ 7.86-7.71 (m, 2H), 7.58 (dd, J=8.3, 1.2 Hz, 1H), 7.57 (dd, J=7.7, 1.2 Hz, 1H), 7.20 (br s, 2H), 5.24 (td, J=7.9, 5.8 Hz, 1H), 4.20-4.02 (m, 2H), 2.98-2.88 (m, 2H), 2.08-1.89 (m, 4H), 0.98 (t, J=7.3 Hz, 3H). ES/MS 428.2 (M+H+).
  • (S)-3-(3-aminopropyl)-5-chloro-2-(cyclopropyl(pyrido[3,2-d]pyrimidin-4-ylamino)methyl)quinazolin-4(3H)-one (Compound 37). 1H NMR (400 MHz, DMSO-d6) δ 8.97-8.92 (m, 1H), 8.86 (br s, 1H), 8.65-8.60 (m, 1H), 8.23-8.18 (m, 1H), 7.98-7.92 (m, 1H), 7.78-7.67 (m, 3H), 7.60 (dd, J=8.2, 1.2 Hz, 1H), 7.57 (dd, J=7.8, 1.2 Hz, 1H), 5.40 (t, J=7.8 Hz, 1H), 4.37-4.27 (m, 1H), 4.19-4.09 (m, 1H), 3.01-2.89 (m, 2H), 2.24-1.95 (m, 2H), 1.81-1.69 (m, 1H), 0.68-0.59 (m, 3H), 0.52-0.44 (m, 1H). ES/MS 436.1 (M+H+).
  • (S)-3-(3-aminopropyl)-5-chloro-2-(cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)quinazolin-4(3H)-one (Compound 38). 1H NMR (400 MHz, DMSO-d6) δ 7.83-7.71 (m, 3H), 7.68-7.55 (m, 4H), 7.45 (br s, 2H), 5.02 (t, J=8.1 Hz, 1H), 4.13-3.91 (m, 2H), 2.96-2.82 (m, 2H), 2.06-1.83 (m, 2H), 1.75-1.62 (m, 1H), 0.65-0.38 (m, 4H). ES/MS 448.8 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-(methylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 39). 1H NMR (400 MHz, DMSO-d6) δ 8.38 (br s, 2H), 7.75 (dd, J=9.1, 6.9 Hz, 1H), 7.59 (dd, J=8.2, 0.9 Hz, 1H), 7.56 (dd, J=7.8, 1.2 Hz, 1H), 7.28 (br s, 1H), 6.94 (br s, 2H), 6.69 (br s, 2H), 5.47-5.32 (m, 1H), 4.20-3.98 (m, 2H), 3.12-2.91 (m, 2H), 2.62-2.55 (m, 3H), 2.15-1.94 (m, 2H), 1.52 (d, J=6.5 Hz, 3H). ES/MS 428.2 (M+H+).
  • (S)-4-amino-6-(((3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 40). 1H NMR (400 MHz, DMSO-d6) δ 8.06 (s, 1H), 7.75 (dd, J=8.2, 7.8 Hz, 1H), 7.72 (br s, 2H), 7.59 (dd, J=8.2, 1.2 Hz, 1H), 7.57 (dd, J=7.8, 1.2 Hz, 1H), 7.49 (br s, 1H), 5.11 (t, J=7.8 Hz, 1H), 4.17-3.99 (m, 2H), 2.95-2.80 (m, 2H), 2.14-1.89 (m, 2H), 1.70-1.58 (m, 1H), 0.63-0.36 (m, 4H). ES/MS 425.1 (M+H+).
  • (S)-2-amino-4-((1-(5-chloro-3-(3-(methylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-6-methylpyrimidine-5-carbonitrile (Compound 41). 1H NMR (400 MHz, DMSO-d6) δ 8.39 (br s, 2H), 7.93 (br s, 1H), 7.75 (dd, J=8.2, 7.8 Hz, 1H), 7.58 (dd, J=8.2, 1.2 Hz, 1H), 7.56 (dd, J=7.7, 1.2 Hz, 1H), 5.39 (p, J=6.4 Hz, 1H), 4.15-3.99 (m, 2H), 3.10-2.92 (m, 2H), 2.58 (t, J=5.4 Hz, 3H), 2.30 (s, 3H), 2.13-1.99 (m, 2H), 1.56 (d, J=6.5 Hz, 3H). ES/MS 427.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 42). 1H NMR (400 MHz, DMSO-d6) δ 7.84-7.71 (m, 4H), 7.59 (dd, J=8.2, 1.1 Hz, 1H), 7.56 (dd, J=7.8, 1.2 Hz, 1H), 7.40 (br s, 2H), 7.08 (br s, 2H), 5.40 (p, J=6.6 Hz, 1H), 4.18-4.00 (m, 2H), 2.98-2.85 (m, 2H), 2.10-1.91 (m, 2H), 1.53 (d, J=6.6 Hz, 3H). ES/MS 414.1 (M+H+).
  • (S)-3-(3-aminopropyl)-2-(((5-bromo-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)(cyclopropyl)methyl)-5-chloroquinazolin-4(3H)-one (Compound 43). 1H NMR (400 MHz, DMSO-d6) δ 12.15 (d, J=2.4 Hz, 1H), 8.20 (s, 1H), 7.74 (dd, J=8.2, 7.8 Hz, 1H), 7.73 (br s, 2H), 7.64 (dd, J=8.2, 1.2 Hz, 1H), 7.57 (dd, J=7.8, 1.2 Hz, 1H), 7.46 (d, J=2.6 Hz, 1H), 7.28 (d, J=7.9 Hz, 1H), 5.72 (dd, J=7.9, 6.2 Hz, 1H), 4.46-4.36 (m, 1H), 4.17-4.07 (m, 1H), 3.04-2.93 (m, 2H), 2.26-2.13 (m, 1H), 2.10-1.97 (m, 1H), 1.59 (q, J=7.1 Hz, 1H), 0.61-0.51 (m, 3H), 0.42-0.34 (m, 1H). ES/MS 504.0 (M+H+).
  • (S)-2-amino-4-(((3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)-6-chloropyrimidine-5-carbonitrile (Compound 44). 1H NMR (400 MHz, DMSO-d6) δ 7.98 (d, J=7.7 Hz, 1H), 7.84 (br s, 1H), 7.77 (t, J=8.0 Hz, 1H), 7.74 (br s, 2H), 7.63 (dd, J=8.2, 1.2 Hz, 1H), 7.59 (dd, J=7.8, 1.2 Hz, 1H), 7.38 (br s, 1H), 5.01 (t, J=8.0 Hz, 1H), 4.08 (ddd, J=14.9, 9.5, 5.8 Hz, 1H), 3.92 (ddd, J=14.5, 9.5, 5.7 Hz, 1H), 2.92-2.82 (m, 2H), 2.04-1.85 (m, 2H), 1.77-1.67 (m, 1H), 0.63-0.55 (m, 2H), 0.55-0.46 (m, 1H), 0.46-0.37 (m, 1H). ES/MS 458.8 (M+H+).
  • (S)-3-(3-aminopropyl)-5-chloro-2-(cyclopropyl(thiazolo[5,4-d]pyrimidin-7-ylamino)methyl)quinazolin-4(3H)-one (Compound 45). 1H NMR (400 MHz, DMSO-d6) δ 9.35 (s, 1H), 8.52 (d, J=7.4 Hz, 1H), 8.46 (s, 1H), 7.73 (dd, J=8.2, 7.8 Hz, 1H), 7.71 (br s, 2H), 7.59 (dd, J=8.2, 1.2 Hz, 1H), 7.56 (dd, J=7.8, 1.2 Hz, 1H), 5.23 (t, J=7.9 Hz, 1H), 4.28-4.08 (m, 2H), 2.96-2.85 (m, 2H), 2.23-1.94 (m, 2H), 1.81-1.64 (m, 1H), 0.65-0.55 (m, 3H), 0.53-0.45 (m, 1H). ES/MS 470.1 (M+H+).
  • (S)-2,4-diamino-6-(((5-chloro-3-(3-(methylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 46). 1H NMR (400 MHz, DMSO-d6) δ 8.43-8.30 (m, 2H), 7.77 (dd, J=8.2, 7.8 Hz, 1H), 7.62 (dd, J=8.2, 1.2 Hz, 1H), 7.58 (dd, J=7.8, 1.2 Hz, 1H), 7.30 (br s, 1H), 7.22 (br s, 2H), 6.94 (br s, 2H), 5.08 (t, J=8.0 Hz, 1H), 4.11-3.95 (m, 2H), 3.04-2.89 (m, 2H), 2.57 (t, J=5.4 Hz, 3H), 2.09-1.88 (m, 2H), 1.73-1.61 (m, 1H), 0.61-0.53 (m, 2H), 0.51-0.36 (m, 2H). ES/MS 454.1 (M+H+).
  • (S)-2,4-diamino-6-(((3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 47). 1H NMR (400 MHz, DMSO-d6) δ 7.77 (dd, J=8.2, 7.8 Hz, 1H), 7.73 (br s, 2H), 7.62 (dd, J=8.2, 1.2 Hz, 1H), 7.58 (dd, J=7.8, 1.2 Hz, 1H), 7.37 (br s, 4H), 7.05 (br s, 2H), 5.07 (t, J=8.0 Hz, 1H), 4.15-3.93 (m, 2H), 2.95-2.83 (m, 2H), 2.06-1.84 (m, 2H), 1.74-1.61 (m, 1H), 0.62-0.52 (m, 2H), 0.52-0.37 (m, 2H). ES/MS 440.2 (M+H+).
  • (S)-3-(3-aminopropyl)-5-chloro-2-(((6-chloropyrido[3,2-d]pyrimidin-4-yl)amino)(cyclopropyl)methyl)quinazolin-4(3H)-one (Compound 48). 1H NMR (400 MHz, DMSO-d6) δ 8.62 (d, J=7.6 Hz, 1H), 8.59 (s, 1H), 8.23 (d, J=8.8 Hz, 1H), 7.97 (d, J=8.8 Hz, 1H), 7.75 (dd, J=8.2, 7.8 Hz, 1H), 7.71 (s, 2H), 7.61 (dd, J=8.2, 1.2 Hz, 1H), 7.57 (dd, J=7.8, 1.2 Hz, 1H), 5.30 (t, J=7.9 Hz, 1H), 4.30-4.20 (m, 1H), 4.17-4.07 (m, 1H), 2.97-2.85 (m, 2H), 2.20-1.94 (m, 2H), 1.83-1.72 (m, 1H), 0.68-0.57 (m, 3H), 0.55-0.45 (m, 1H). ES/MS 470.1 (M+H+).
  • (S)-2-amino-4-(((3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)-6-methylpyrimidine-5-carbonitrile (Compound 49). 1H NMR (400 MHz, DMSO-d6) δ 7.76 (dd, J=8.2, 7.8 Hz, 1H), 7.72 (br s, 2H), 7.62 (dd, J=8.2, 1.2 Hz, 1H), 7.58 (dd, J=7.8, 1.2 Hz, 1H), 7.56 (br s, 1H), 7.41 (br s, 1H), 7.00 (br s, 1H), 5.08 (t, J=8.0 Hz, 1H), 4.13-3.91 (m, 2H), 2.86 (d, J=7.3 Hz, 2H), 2.29 (s, 3H), 2.05-1.85 (m, 2H), 1.76-1.64 (m, 1H), 0.64-0.52 (m, 2H), 0.52-0.35 (m, 2H). ES/MS 439.1 (M+H+).
  • (S)-3,5-diamino-N-(1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)-6-bromopyrazine-2-carboxamide (Compound 50). 1H NMR (400 MHz, DMSO-d6) δ 8.34 (d, J=7.5 Hz, 1H), 7.80-7.69 (m, 4H), 7.61-7.52 (m, 2H), 7.04 (s, 2H), 5.21 (p, J=6.7 Hz, 1H), 4.27 (ddd, J=14.3, 8.7, 5.9 Hz, 1H), 4.03 (ddd, J=14.5, 8.9, 6.2 Hz, 1H), 2.94 (q, J=6.3 Hz, 2H), 2.13-1.91 (m, 2H), 1.53 (d, J=6.5 Hz, 3H). ES/MS 497.1 (M+H+).
  • (S)-3-(3-aminopropyl)-5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)quinazolin-4(3H)-one (Compound 71);
  • (S)-3-(3-aminopropyl)-5-chloro-2-(1-(thiazolo[5,4-d]pyrimidin-7-ylamino)ethyl)quinazolin-4(3H)-one (Compound 72);
  • (S)-4-amino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 73);
  • (S)-4-amino-6-((1-(3-(3-aminocyclobutyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 86). 1H NMR (400 MHz, DMSO-d6) δ 8.10 (s, 1H), 7.72 (m, 1H), 7.54 (m, 2H), 7.38 (m, 1H), 5.53 (m, 1H), 4.95 (m, 1H), 4.11 (m, 1H), 3.08 (m, 2H), 2.44 (m, 1H), 2.28 (m, 1H), 1.45 (d, J=8 Hz, 3H). ES/MS 411.0 (M+H+).
  • (S)-4-amino-6-((1-(5-chloro-4-oxo-3-(piperidin-4-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 87). 1H NMR (400 MHz, DMSO-d6) δ 8.46 (m, 1H), 8.07 (s, 1H), 7.69 (m, 1H), 7.52 (m, 2H), 7.38 (m, 2H), 5.60 (m, 1H), 4.30 (m, 2H), 3.37 (m, 1H), 3.05 (m, 3H), 2.61 (m, 1H), 1.97 (m, 2H), 1.49 (d, J=6.5 Hz, 3H). ES/MS 439.0 (M+H+).
  • 4-amino-6-(((1S)-1-(5-chloro-3-(morpholin-2-ylmethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 88). 1H NMR (400 MHz, DMSO-d6) δ 8.89 (s, 1H), 8.74 (s, 1H), 8.07 (s, 1H), 7.74 (m, 1H), 7.54 (m, 2H), 7.35 (m, 2H), 5.54 (m, 1H), 4.19 (m, 2H), 4.07 (m, 1H), 3.90 (m, 1H), 3.57 (m, 1H), 3.41 (m, 1H), 3.16 (m, 1H), 3.00 (m, 1H), 2.85 (m, 1H), 1.49 (d, J=6.5 Hz, 3H). ES/MS 441.2 (M+H+).
  • 4-amino-6-(((1S)-1-(5-chloro-4-oxo-3-(pyrrolidin-3-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 89). 1H NMR (400 MHz, DMSO-d6) δ 8.63 (s, 2H), 7.98 (s, 1H), 7.69 (m, 1H), 7.49 (m, 2H), 7.34 (m, 1H), 5.34 (m, 1H), 4.25 (m, 1H), 3.97 (m, 1H), 3.25 (m, 2H), 2.93 (m, 2H), 2.88 (m, 1H), 2.01 (m, 1H), 1.76 (m, 1H), 1.47 (d, J=6.5 Hz, 3H). ES/MS 425.1 (M+H+).
  • 4-amino-6-(((1S)-1-(5-chloro-4-oxo-3-(pyrrolidin-2-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 90). 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 8.04 (s, 1H), 7.75 (m, 1H), 7.56 (m, 2H), 7.43 (m, 1H), 5.42 (m, 1H), 4.64 (m, 1H), 4.36 (m, 2H), 4.14 (m, 1H), 3.91 (m, 1H), 3.34 (m, 1H), 3.10 (m, 1H), 2.16 (m, 1H), 1.73 (m, 1H), 1.52 (d, J=6.5 Hz, 3H). ES/MS 425.1 (M+H+).
  • 4-amino-6-(((1S)-1-(5-chloro-4-oxo-3-(piperidin-2-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 91). 1H NMR (400 MHz, DMSO-d6) δ 8.60 (m, 1H), 8.37 (m, 1H), 8.07 (s, 1H), 7.70 (m, 1H), 7.55 (m, 2H), 7.43 (m, 1H), 5.24 (m, 1H), 4.63 (m, 1H), 3.94 (m, 1H), 3.84 (m, 1H), 3.29 (m, 1H), 2.67 (m, 1H), 2.02 (m, 1H), 1.78 (m, 1H), 1.68 (m, 2H), 1.51 (d, J=6.5 Hz, 3H), 1.43 (m, 1H). ES/MS 439.2 (M+H+).
  • 4-amino-6-(((1S)-1-(5-chloro-4-oxo-3-(piperidin-3-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 92). 1H NMR (400 MHz, DMSO-d6) δ 8.56 (m, 1H), 8.08 (s, 1H), 7.70 (m, 1H), 7.54 (m, 2H), 7.39 (m, 1H), 5.35 (m, 1H), 4.14 (m, 1H), 3.82 (m, 1H), 3.21 (m, 2H), 2.78 (m, 2H), 2.38 (m, 1H), 1.81 (m, 2H), 1.57 (m, 1H), 1.52 (d, J=6.5 Hz, 3H), 1.38 (m, 1H). ES/MS 439.3 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5-chloro-4-oxo-3-((R)-piperidin-3-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 123). 1H NMR (400 MHz, DMSO-d6) δ 8.53 (d, J=11.3 Hz, 1H), 8.12 (d, J=11.5 Hz, 1H), 7.62-7.50 (m, 2H), 7.40 (s, 1H), 7.04 (d, J=81.1 Hz, 4H), 5.38 (q, J=6.9 Hz, 1H), 4.11 (dd, J=14.5, 8.7 Hz, 1H), 3.83 (dd, J=14.4, 5.8 Hz, 1H), 3.25-3.14 (m, 3H), 2.80 (q, J=11.8 Hz, 1H), 2.73-2.62 (m, 1H), 2.14 (d, J=11.7 Hz, 1H), 1.77 (d, J=15.4 Hz, 2H), 1.66-1.52 (m, 1H), 1.49 (d, J=6.4 Hz, 3H), 1.34 (qd, J=12.7, 12.1, 4.4 Hz, 1H). ES/MS 454.2 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5-chloro-4-oxo-3-((S)-piperidin-3-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 124). 1H NMR (400 MHz, DMSO-d6) δ 8.59 (d, J=11.4 Hz, 1H), 8.11 (d, J=11.5 Hz, 1H), 7.79-7.67 (m, 1H), 7.63-7.50 (m, 2H), 7.39-7.26 (m, 1H), 7.16 (s, 2H), 7.06-6.81 (m, 2H), 5.54-5.40 (m, 1H), 3.95 (dq, J=15.8, 7.3 Hz, 2H), 3.20 (t, J=16.3 Hz, 2H), 2.77 (dt, J=21.9, 10.9 Hz, 2H), 2.15 (s, 1H), 1.82-1.65 (m, 2H), 1.47 (d, J=6.5 Hz, 3H), 1.29 (td, J=12.8, 12.4, 9.2 Hz, 1H). ES/MS 454.2 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5-chloro-4-oxo-3-((S)-pyrrolidin-3-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 125). 1H NMR (400 MHz, DMSO-d6) δ 8.75-8.50 (m, 2H), 7.72 (t, J=8.0 Hz, 1H), 7.60-7.46 (m, 2H), 7.35 (s, 1H), 7.09 (s, 1H), 6.84 (s, 1H), 5.41 (p, J=6.8 Hz, 1H), 4.17 (dd, J=14.7, 8.1 Hz, 1H), 4.08 (dd, J=14.6, 5.8 Hz, 1H), 3.73 (t, J=5.3 Hz, 1H), 3.13-2.97 (m, 2H), 2.74-2.60 (m, 1H), 2.10 (dtd, J=11.7, 7.2, 4.2 Hz, 1H), 1.69 (dt, J=12.8, 8.8 Hz, 1H), 1.61 (q, J=5.9, 4.5 Hz, 1H), 1.56-1.52 (m, 1H), 1.49 (d, J=6.5 Hz, 3H). ES/MS 440.2 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5-chloro-4-oxo-3-((R)-pyrrolidin-3-ylmethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 126). 1H NMR (400 MHz, DMSO-d6) δ 8.66 (d, J=23.6 Hz, 2H), 7.72 (t, J=8.0 Hz, 1H), 7.55 (ddd, J=12.7, 8.0, 1.2 Hz, 2H), 7.35 (d, J=12.2 Hz, 1H), 7.13 (s, 2H), 6.87 (s, 2H), 5.44 (p, J=6.7 Hz, 1H), 4.24-4.03 (m, 2H), 3.12-2.90 (m, 2H), 2.74-2.57 (m, 1H), 1.99 (dtd, J=12.4, 7.3, 4.8 Hz, 1H), 1.73 (dq, J=12.9, 8.4 Hz, 1H), 1.48 (d, J=6.5 Hz, 3H). ES/MS 440.2 (M+H+).

G. Preparation of a compound of formula (I) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, R2 is NHCOCH3, R3 is methyl, R4 is 4-amino-5-cyanopyrimidin-2-yl, and R5 is hydrogen

To (S)-4-amino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (200 mg, 0.502 mmol), HATU (285 mg, 0.75 mmol) and acetic acid (45 mg, 0.75 mmol) in DMF (2 mL) was added Hunig's base (0.3 mL). After stirring at room temp for 1 hour, water was added and the mixture was extracted with EtOAc (3×20 mL), dried (MgSO4), and concentrated. The residue was purified on a reverse phase HPLC run from 0 to 95% ACN in water (0.1% TFA) to give (S)—N-(3-(2-(1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)acetamide (Compound 51). 1H NMR (400 MHz, DMSO-d6) δ 8.06 (s, 1H), 7.94 (t, J=5.8 Hz, 1H), 7.87 (d, J=6.9 Hz, 1H), 7.71 (t, J=8.0 Hz, 1H), 7.54 (ddd, J=7.8, 5.5, 1.2 Hz, 2H), 7.48 (s, 2H), 5.42 (p, J=6.7 Hz, 1H), 4.13 (ddd, J=14.5, 9.9, 5.7 Hz, 1H), 3.89 (ddd, J=14.5, 10.1, 5.6 Hz, 1H), 3.11 (q, J=6.5 Hz, 2H), 1.88 (ddt, J=23.2, 10.5, 7.0 Hz, 2H), 1.80 (s, 3H), 1.54 (d, J=6.6 Hz, 3H). ES/MS 441.2 (M+H+).

H. Preparation of the below compounds of formula (I), using Example 3G and Reaction Scheme I:

  • (S)—N-(3-(5-chloro-2-(cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)pivalamide (Compound 52). 1H NMR (400 MHz, DMSO-d6) δ 7.87 (t, J=5.9 Hz, 1H), 7.73 (dd, J=8.3, 7.7 Hz, 1H), 7.55 (ddd, J=7.8, 5.3, 1.2 Hz, 2H), 6.73 (s, 2H), 6.58 (s, 2H), 6.40 (d, J=8.7 Hz, 1H), 5.30 (t, J=7.9 Hz, 1H), 4.19 (t, J=10.6 Hz, 1H), 3.94 (t, J=11.8 Hz, 1H), 3.04 (dd, J=13.3, 7.2 Hz, 1H), 2.05-1.90 (m, 1H), 1.73 (d, J=6.5 Hz, 1H), 1.51 (h, J=7.8 Hz, 2H), 1.12 (s, 9H), 0.60-0.49 (m, 2H), 0.48-0.39 (m, 1H), 0.38-0.26 (m, 1H). ES/MS 524.2 (M+H+).
  • (S)—N-(3-(5-chloro-2-(cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)propyl)acetamide (Compound 53). 1H NMR (400 MHz, DMSO-d6) δ 8.12 (t, J=6.0 Hz, 1H), 7.75 (dd, J=8.3, 7.7 Hz, 1H), 7.58 (ddd, J=10.9, 8.0, 1.2 Hz, 5H), 5.16 (t, J=8.0 Hz, 1H), 4.09-3.85 (m, 2H), 3.22 (dd, J=13.1, 6.7 Hz, 1H), 3.04 (dt, J=13.4, 6.6 Hz, 1H), 1.83 (d, J=0.9 Hz, 3H), 1.76 (s, 1H), 1.69-1.56 (m, 2H), 0.57 (dt, J=8.1, 4.2 Hz, 3H), 0.43 (p, J=4.8 Hz, 3H). ES/MS 482.2 (M+H+).

I. Preparation of a compound of formula (I) wherein n is 1, R1 is chloro, m is 0, R3 is methyl, B is (CH2)3, R2 is NHoxetanyl, R3 is cyclopropyl, R4 is 2,6-diamino-5-cyanopyrimidin-2-yl, and R5 is hydrogen

To (S)-2,4-diamino-6-(((3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (100 mg, 0.23 mmol) in MeOH (5 mL), was added 3-oxetanone (82 mg, 1.15 mmol). After stirring at room temp for 12 hour, sodium triacetoxyborohydride (964 mg, 4.5 mmol) was added. After stirring for 3 hour at room temperature saturated sodium bicarbonate solution (30 mL) was added and the mixture was extracted with EtOAc (3×30 mL) and concentrated. The residue was purified on a reverse phase HPLC run from 0 to 95% ACN in water (0.1% TFA) to (S)-2,4-diamino-6-(((5-chloro-3-(3-(oxetan-3-ylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 55). 1H NMR (400 MHz, DMSO-d6) δ 9.25 (s, 2H), 7.83-7.69 (m, 1H), 7.67-7.47 (m, 2H), 7.12 (s, 2H), 6.84 (s, 2H), 5.10 (t, J=7.9 Hz, 1H), 4.73 (dd, J=8.2, 6.7 Hz, 2H), 4.60 (q, J=6.7 Hz, 2H), 4.35 (d, J=7.0 Hz, 1H), 4.06 (t, J=7.5 Hz, 2H), 2.95 (s, 2H), 1.99 (s, 2H), 1.74-1.58 (m, 1H), 0.57 (dd, J=8.4, 4.2 Hz, 2H), 0.51-0.31 (m, 2H). ES/MS 496.2 (M+H+).

J. Preparation of the below compounds of formula (I), using Example 31 and Reaction Scheme I:

  • (S)-2,4-diamino-6-(((5-chloro-3-(3-(cyclobutylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 54). 1H NMR (400 MHz, DMSO-d6) δ 8.65 (s, 2H), 7.88-7.70 (m, 1H), 7.66-7.53 (m, 2H), 7.18 (d, J=34.9 Hz, 2H), 6.88 (s, 2H), 5.09 (t, J=8.0 Hz, 1H), 4.03 (t, J=7.7 Hz, 2H), 3.72-3.57 (m, 1H), 2.99-2.74 (m, 2H), 2.23-2.05 (m, 3H), 1.98 (dq, J=16.0, 8.1, 7.6 Hz, 2H), 1.78 (td, J=9.9, 7.1 Hz, 2H), 1.67 (dq, J=8.0, 4.1, 3.0 Hz, 1H), 1.32-1.13 (m, 1H), 0.56 (dd, J=8.2, 4.6 Hz, 2H), 0.50-0.43 (m, 1H), 0.43-0.33 (m, 1H). ES/MS 494.2 (M+H+).
  • (S)-2,4-diamino-6-(((5-chloro-3-(3-((4,4-difluorocyclohexyl)amino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 56). 1H NMR (400 MHz, DMSO-d6) δ 8.47 (d, J=47.0 Hz, 2H), 7.85-7.70 (m, 1H), 7.68-7.51 (m, 2H), 7.20 (s, 2H), 6.93 (s, 1H), 5.13 (t, J=8.0 Hz, 1H), 4.06 (t, J=7.5 Hz, 2H), 3.18 (s, 1H), 3.01 (s, 2H), 2.21-1.75 (m, 9H), 1.67 (dtd, J=13.2, 8.1, 5.1 Hz, 1H), 1.61-1.44 (m, 2H), 0.62-0.52 (m, 2H), 0.52-0.42 (m, 1H), 0.42-0.30 (m, 1H). ES/MS 558.2 (M+H+).

K. Preparation of a compound of formula (I) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, R2 is NH(pyrimidin-2-yl), R3 is methyl, R4 is 2,6-diamino-5-cyanopyrimidin-2-yl, and R5 is hydrogen

A solution of (S)-2,4-diamino-6-((1-(3-(3-aminopropyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (80 mg, 0.19 mmol, 1 equiv) and 2-chloropyrimidine (22 mg, 0.193) in isopropyl alcohol (2 mL) was stirred at 85° C. for seven days. The reaction was concentrated and purified by Prep HPLC to give (S)-2,4-diamino-6-((1-(5-chloro-4-oxo-3-(3-(pyrimidin-2-ylamino)propyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 4). 1H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J=4.8 Hz, 2H), 8.04 (br s, 2H), 7.81 (br s, 2H), 7.74 (t, J=8.0 Hz, 1H), 7.57 (ddd, J=9.1, 8.0, 1.2 Hz, 2H), 7.32 (br s, 2H), 6.59 (t, J=4.8 Hz, 1H), 5.50-5.37 (m, 1H), 4.11-3.97 (m, 2H), 3.41-3.32 (m, 2H), 2.01-1.90 (m, 1H), 1.50 (d, J=6.5 Hz, 3H). ES/MS 492.2 (M+H+).

L. Preparation of the below compounds of formula (I), using Example 3K and Reaction Scheme I:

  • (S)-2,4-diamino-6-((1-(5-chloro-4-oxo-3-(3-(pyridin-2-ylamino)propyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 5). 1H NMR (400 MHz, DMSO-d6) δ 7.99 (d, J=23.6 Hz, 4H), 7.74 (t, J=8.2 Hz, 1H), 7.58 (dd, J=8.2, 1.2 Hz, 1H), 7.55 (dd, J=7.8, 1.1 Hz, 1H), 7.36 (s, 2H), 7.05 (s, 2H), 5.42 (p, J=7.0 Hz, 1H), 4.04 (t, J=7.8 Hz, 1H), 3.60-3.46 (m, 1H), 3.45-3.33 (m, 1H), 2.03-1.88 (m, 2H), 1.54 (s, 3H), 1.52 (s, 3H). ES/MS 490.2 (M+H+).

M. Preparation of a compound of formula (I) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, R2 is N(CH3)2, R3 is methyl, R4 is 4-amino-5-cyanopyrimidin-2-yl, and R5 is hydrogen

To (S)-2-(1-aminoethyl)-5-chloro-3-(3-(dimethylamino)propyl)quinazolin-4(3H)-one (200 mg, 0.65 mmol) and 4-amino-6-chloropyrimidine-5-carbonitrile (100 mg, 0.65 mmol) in IPA (2 mL) was added Hunig's base (0.3 mL). After stirring at 90° C. for 4 hours in a microwave reactor, water was added and the mixture was extracted with EtOAc (3×50 mL), dried (MgSO4), and concentrated. The residue was purified on a reverse phase system run from 0 to 95% ACN in water (0.1% TFA) to give (S)-4-amino-6-((1-(5-chloro-3-(3-(dimethylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 57). 1H NMR (400 MHz, DMSO-d6) δ 8.18 (s, 1H), 8.16 (d, J=6.1 Hz, 1H), 7.98 (s, 1H), 7.73 (dd, J=8.5, 7.5 Hz, 2H), 7.65 (s, 1H), 7.59-7.51 (m, 1H), 5.42 (p, J=6.7 Hz, 1H), 4.23 (ddd, J=14.4, 8.8, 5.8 Hz, 1H), 4.01 (ddd, J=14.5, 8.7, 6.1 Hz, 1H), 2.91 (dd, J=10.5, 4.4 Hz, 2H), 2.58 (s, 6H) 2.11 (ddq, J=43.7, 14.1, 6.6 Hz, 2H), 1.56 (d, J=6.6 Hz, 3H). ES/MS 427.2 (M+H+).

N. Preparation of the below compounds of formula (I), using Example 3M and Reaction Scheme I:

  • (S)-2,4-diamino-6-((1-(5-chloro-3-(2-(dimethylamino)ethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 58). 1H NMR (400 MHz, DMSO-d6) δ 8.37 (d, J=10.8 Hz, 2H), 7.75 (dd, J=9.0, 7.0 Hz, 1H), 7.57 (m, 1H), 7.29 (d, J=11.9 Hz, 1H), 6.94 (s, 2H), 6.68 (s, 2H), 5.39 (q, J=6.9 Hz, 1H), 4.06 (dd, J=21.6, 14.1 Hz, 3H), 2.40 (s, 6H), 2.06 (d, J=13.5 Hz, 2H), 1.52 (d, J=6.4 Hz, 2H). ES/MS 428.2 (M+H+).
  • (S)-2-(1-((9H-purin-6-yl)amino)ethyl)-5-chloro-3-(3-morpholinopropyl)quinazolin-4(3H)-one (Compound 19). 1H NMR (400 MHz, DMSO-d6) δ 9.79 (s, 1H), 8.45 (s, 1H), 8.38-8.25 (m, 2H), 7.68 (t, J=8.0 Hz, 1H), 7.56-7.47 (m, 1H), 5.63-5.46 (m, 1H), 4.38-4.22 (m, 1H), 4.10-3.90 (m, 3H), 3.70-3.55 (m, 2H), 3.41 (t, J=15.2 Hz, 2H), 3.30-3.18 (m, 2H), 3.13-2.96 (m, 2H), 2.39-2.26 (m, 1H), 2.25-2.12 (m, 1H), 1.63 (d, J=6.5 Hz, 3H). ES/MS 469.1 (M+H+).
  • 2,4-diamino-6-(((1S)-1-(5-chloro-3-(3-hydroxy-4-(isopropylamino)butyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 74). 1H NMR (400 MHz, DMSO-d6) δ 8.27 (s, 2H), 7.79-7.67 (m, 1H), 7.62-7.50 (m, 3H), 7.27-7.21 (m, 2H), 6.94 (s, 2H), 5.55 (s, 1H), 5.44 (q, J=6.8 Hz, 1H), 4.11 (t, J=7.8 Hz, 1H), 3.91-3.77 (m, 1H), 3.33-3.24 (m, 1H), 3.05-2.97 (m, 1H), 2.86-2.81 (m, 1H), 1.90 (q, J=7.3 Hz, 2H), 1.60-1.49 (m, 3H), 1.28-1.17 (m, 6H). ES/MS 486.2 (M+H+).
  • 2,4-diamino-6-(((1S)-1-(3-(4-(tert-butylamino)-3-hydroxybutyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 75). 1H NMR (400 MHz, DMSO-d6) δ 8.32-8.24 (m, 1H), 8.23-8.18 (m, 1H), 7.80-7.64 (m, 2H), 7.63-7.51 (m, 2H), 7.34 (s, 2H), 7.03 (s, 2H), 5.63-5.39 (m, 2H), 4.38-3.93 (m, 1H), 3.79 (d, J=10.3 Hz, 1H), 3.01 (d, J=10.6 Hz, 1H), 2.81-2.71 (m, 1H), 1.98-1.88 (m, 2H), 1.61-1.51 (m, 3H), 1.27 (s, 9H). ES/MS 500.2 (M+H+).
  • (S)-5-(5-chloro-2-(cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)pentanamide (Compound 59). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (dd, J=8.2, 7.8 Hz, 1H), 7.58 (dd, J=8.2, 1.2 Hz, 1H), 7.54 (dd, J=7.8, 1.2 Hz, 1H), 7.29 (br s, 1H), 6.77 (br s, 1H), 6.17 (d, J=8.7 Hz, 1H), 6.12 (br s, 2H), 5.77 (br s, 2H), 5.20 (dd, J=8.8, 7.4 Hz, 1H), 4.15-3.86 (m, 2H), 2.16-2.05 (m, 2H), 1.78-1.44 (m, 5H), 0.57-0.32 (m, 4H). ES/MS 491.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(3-(3-(benzyl(methyl)amino)propyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 60). 1H NMR (400 MHz, DMSO-d6) δ 7.88-6.98 (m, 8H), 6.82-6.57 (m, 2H), 6.43 (br s, 2H), 5.55-5.30 (m, 1H), 4.51-3.77 (m, 3H), 3.55-2.98 (m, 2H), 2.75-2.57 (m, 1H), 2.46-1.82 (m, 5H), 1.51 (d, J=6.3 Hz, 3H). ES/MS 518.2 (M+H+).
  • (S)-2-amino-4-((1-(3-(3-(benzyl(methyl)amino)propyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-6-methylpyrimidine-5-carbonitrile (Compound 61). 1H NMR (400 MHz, DMSO-d6) δ 9.49 (br s, 1H), 7.83-7.11 (m, 8H), 6.85 (br s, 1H), 5.59-5.27 (m, 1H), 4.52-3.78 (m, 4H), 3.59-3.03 (m, 2H), 2.77-2.62 (m, 2H), 2.46-1.81 (m, 6H), 1.55 (d, J=6.3 Hz, 3H). ES/MS 517.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-(methyl(2,2,2-trifluoroethyl)amino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 62). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (t, J=8.1 Hz, 1H), 7.57 (dd, J=8.2, 1.2 Hz, 1H), 7.53 (dd, J=7.8, 1.2 Hz, 1H), 7.07 (d, J=7.8 Hz, 1H), 6.64 (br s, 2H), 5.52-5.40 (m, 1H), 4.18-4.02 (m, 1H), 3.94-3.78 (m, 1H), 3.23-3.09 (m, 2H), 2.62-2.53 (m, 2H), 2.35 (s, 3H), 1.99-1.80 (m, 2H), 1.50 (d, J=6.5 Hz, 3H). ES/MS 510.1 (M+H+).
  • (S)-3-(5-chloro-2-(cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)propanenitrile (Compound 63). 1H NMR (DMSO-d6) δ 7.78 (t, J=8.0 Hz, 1H), 7.64 (dd, J=8.0, 0.9 Hz, 1H), 7.60 (dd, J=7.7, 1.1 Hz, 1H), 5.04 (t, J=8.1 Hz, 1H), 4.36-4.22 (m, 1H), 4.17-4.05 (m, 1H), 3.14-3.01 (m, 1H), 2.85-2.72 (m, 1H), 1.74-1.62 (m, 1H), 0.66-0.41 (m, 4H). ES/MS 445.1 (M+1+).
  • (S)-5-(5-chloro-2-(cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)pentanamide (Compound 64). 1H NMR (400 MHz, DMSO-d6) δ 7.73 (t, J=8.0 Hz, 1H), 7.59 (dd, J=8.2, 1.2 Hz, 1H), 7.55 (dd, J=7.8, 1.2 Hz, 1H), 7.29 (br s, 1H), 6.79 (br s, 1H), 6.77 (d, J=8.3 Hz, 1H), 6.69 (br s, 2H), 6.53 (br s, 2H), 5.21 (t, J=8.1 Hz, 1H), 3.96 (t, J=7.5 Hz, 2H), 2.18-2.02 (m, 2H), 1.73-1.43 (m, 5H), 0.61-0.39 (m, 3H), 0.40-0.28 (m, 1H). ES/MS 482.2 (M+H+).
  • (S)-4-(5,8-dichloro-2-(cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)butanamide (Compound 65). 1H NMR (400 MHz, DMSO-d6) δ 7.91 (d, J=8.5 Hz, 1H), 7.52 (d, J=8.5 Hz, 1H), 7.44 (br s, 1H), 6.92 (br s, 1H), 6.73 (br s, 2H), 6.58 (br s, 2H), 6.27 (d, J=8.7 Hz, 1H), 5.48 (dd, J=8.5, 7.2 Hz, 2H), 4.28-4.16 (m, 1H), 3.97-3.86 (m, 1H), 2.29-2.15 (m, 3H), 1.94-1.78 (m, 1H), 1.60-1.43 (m, 1H), 0.62-0.45 (m, 3H), 0.37-0.27 (m, 1H). ES/MS 502.1 (M+H+).
  • (S)-4-(5-chloro-2-(cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)butanamide (Compound 66). 1H NMR (400 MHz, DMSO-d6) δ 7.73 (t, J=8.0 Hz, 1H), 7.59-7.53 (m, 2H), 7.46 (br s, 1H), 6.93 (br s, 1H), 5.33-5.21 (m, 1H), 4.27-4.08 (m, 1H), 4.02-3.83 (m, 1H), 2.27-2.07 (m, 3H), 1.92-1.75 (m, 1H), 1.64-1.49 (m, 1H), 0.62-0.46 (m, 3H), 0.42-0.30 (m, 1H). ES/MS 468.2 (M+H+).
  • 2-[3-[5-chloro-2-[(S)-cyclopropyl-[(2,6-diamino-5-chloro-pyrimidin-4-yl)amino]methyl]-4-oxo-quinazolin-3-yl]phenyl]acetonitrile.
  • (S)-4-((1-(3-((1H-pyrazol-3-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-2,6-diaminopyrimidine-5-carbonitrile (Compound 76). 1H NMR (400 MHz, DMSO-d6) δ 12.68 (s, 1H), 7.81-7.59 (m, 2H), 7.52 (ddd, J=8.0, 1.2 Hz, 2H), 6.91 (d, J=6.8 Hz, 1H), 6.61 (s, 2H), 6.39 (br s, 2H), 6.20 (d, J=2.2 Hz, 1H), 5.62-5.16 (m, 3H), 1.25 (d, J=6.5 Hz, 3H). ES/MS 437.1 (M+H+).
  • (S)-4-((1-(3-((1H-pyrazol-3-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-6-aminopyrimidine-5-carbonitrile (Compound 77). 1H NMR (400 MHz, DMSO-d6) δ 12.65 (s, 1H), 7.97 (s, 1H), 7.77-7.67 (m, 1H), 7.63 (br s, 1H), 7.59 (br d, J=6.7 Hz, 1H), 7.57-7.48 (m, 2H), 7.44-7.04 (br m, 2H), 6.17 (d, J=2.2 Hz, 1H), 5.61-5.48 (m, 1H), 5.44-5.19 (m, 2H), 1.35 (d, J=6.5 Hz, 3H). ES/MS 422.1 (M+H+).
  • (S)-3-((1H-pyrazol-3-yl)methyl)-5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)quinazolin-4(3H)-one (Compound 78). 1H NMR (400 MHz, DMSO-d6) δ 12.66 (s, 1H), 9.09 (br s, 1H), 8.14-8.00 (m, 2H), 7.74-7.41 (m, 5H), 6.20 (d, J=2.2 Hz, 1H), 5.76-5.56 (m, 1H), 5.56-5.28 (m, 2H), 1.45 (d, J=6.5 Hz, 3H). ES/MS 422.1 (M+H+).
  • (S)-4-((1-(3-((1H-pyrazol-4-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-2,6-diaminopyrimidine-5-carbonitrile (Compound 79). 1H NMR (400 MHz, DMSO-d6) δ 8.10 (br s, 1H), 7.94-7.23 (br m, 2H), 7.85 (br s, 2H), 7.71 (dd, J=8.3, 7.7 Hz, 1H), 7.62 (s, 2H), 7.57-7.48 (m, 2H), 5.59-5.41 (m, 1H), 5.22 (d, J=15.6 Hz, 1H), 5.01 (d, J=15.6 Hz, 1H), 1.37 (d, J=6.5 Hz, 3H). ES/MS 437.1 (M+H+).
  • (S)-4-((1-(3-((1H-imidazol-2-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-2,6-diaminopyrimidine-5-carbonitrile (Compound 80). 1H NMR (400 MHz, DMSO-d6) δ 7.83-7.76 (m, 1H), 7.65 (dd, J=8.2, 1.2 Hz, 1H), 7.59 (dd, J=7.8, 1.2 Hz, 1H), 7.55 (s, 2H), 7.33-6.81 (br m, 5H), 5.55-5.33 (m, 3H), 1.43 (d, J=6.4 Hz, 3H). ES/MS 437.1 (M+H+).
  • (S)-4-((1-(3-((1H-1,2,4-triazol-3-yl)methyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-2,6-diaminopyrimidine-5-carbonitrile (Compound 81). 1H NMR (400 MHz, DMSO-d6) δ 8.32 (br s, 1H), 8.10 (br s, 1H), 7.84 (br s, 2H), 7.78-7.68 (m, 1H), 7.62-7.49 (m, 2H), 7.60-7.22 (br m, 2H), 5.49 (d, J=16.8 Hz, 1H), 5.41-5.34 (m, 1H), 5.31 (d, J=16.8 Hz, 1H), 1.39 (d, J=6.5 Hz, 3H). ES/MS 438.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(2-cyanoethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 104). 1H NMR (400 MHz, DMSO-d6) δ 7.74 (td, J=8.1, 1.1 Hz, 1H), 7.57 (ddt, J=7.8, 4.5, 1.3 Hz, 2H), 5.54-5.29 (m, 1H), 4.36 (dt, J=14.6, 7.6 Hz, 2H), 4.16 (dt, J=14.1, 6.8 Hz, 2H), 1.52 (d, J=6.5 Hz, 3H). ES/MS 410.1 (M+H+).
  • ((S)-4-amino-6-((1-(5-chloro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 105). 1H NMR (400 MHz, DMSO-d6) δ 8.03 (s, 1H), 7.81 (d, J=7.1 Hz, 1H), 7.69 (t, J=8.0 Hz, 1H), 7.51 (td, J=8.0, 1.2 Hz, 2H), 7.42 (s, 1H), 5.45 (p, J=6.6 Hz, 1H), 4.13 (ddd, J=14.5, 8.7, 6.4 Hz, 1H), 3.84 (dt, J=14.3, 6.9 Hz, 1H), 3.32 (td, J=6.1, 1.9 Hz, 2H), 1.94 (ddt, J=12.8, 9.5, 4.7 Hz, 2H), 1.50 (d, J=6.6 Hz, 3H). ES/MS 414.1 (M+H+).
  • (S)-4-amino-6-((1-(5,8-dichloro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 106). 1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J=0.5 Hz, 1H), 8.00-7.79 (m, 1H), 7.61-7.35 (m, 2H), 5.66-5.36 (m, 1H), 4.09 (dd, J=14.4, 6.7 Hz, 3H), 3.29 (t, J=6.0 Hz, 2H), 3.25-3.04 (m, 3H), 1.90 (s, 2H), 1.52 (d, J=6.5 Hz, 3H). ES/MS 449.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(5,8-dichloro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 107). 1H NMR (400 MHz, DMSO-d6) δ 8.04-7.84 (m, 1H), 7.79-7.26 (m, 3H), 5.73-5.15 (m, 1H), 4.23-3.78 (m, 2H), 3.53-3.28 (m, 2H), 3.32-3.04 (m, 3H), 2.04-1.74 (m, 2H), 1.63-1.31 (m, 3H). ES/MS 464.1 (M+H+).
  • (S)-4-amino-6-((1-(5,8-difluoro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 108). 1H NMR (400 MHz, DMSO-d6) δ 7.79 (d, J=7.1 Hz, 1H), 7.77-7.56 (m, 1H), 7.41 (s, 1H), 7.25 (ddd, J=10.5, 9.0, 3.6 Hz, 2H), 5.48 (p, J=6.7 Hz, 1H), 4.14 (dt, J=14.3, 7.5 Hz, 2H), 3.87 (dt, J=14.4, 7.5 Hz, 2H), 3.31 (td, J=6.2, 1.7 Hz, 2H), 3.15 (s, 3H), 1.94 (p, J=7.0 Hz, 2H), 1.51 (d, J=6.5 Hz, 3H). ES/MS 431.1 (M+H+). ES/MS 416.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(5,8-difluoro-3-(3-methoxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 109). 1H NMR (400 MHz, DMSO-d6) δ 7.88 (s, 1H), 7.81-7.44 (m, 3H), 7.42-6.84 (m, 3H), 5.45 (q, J=6.8 Hz, 1H), 3.91 (dq, J=14.5, 6.9, 6.2 Hz, 4H), 3.22-3.06 (m, 3H), 2.16-1.76 (m, 2H), 1.50 (d, J=6.3 Hz, 3H). ES/MS 431.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-cyanopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 110). 1H NMR (400 MHz, DMSO-d6) δ 7.96 (s, 1H), 7.81-7.58 (m, 2H), 7.53 (ddt, J=8.6, 7.7, 1.0 Hz, 2H), 5.37 (t, J=6.8 Hz, 1H), 3.94 (q, J=7.9, 7.3 Hz, 3H), 2.79-2.55 (m, 2H), 1.99 (tt, J=14.9, 6.5 Hz, 2H), 1.50 (d, J=6.4 Hz, 3H). ES/MS 424.1 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 111). 1H NMR (400 MHz, DMSO-d6) δ 7.84-7.61 (m, 1H), 7.65-7.35 (m, 2H), 7.19 (s, 2H), 5.41 (p, J=6.8 Hz, 1H), 4.21 (ddd, J=14.8, 9.7, 5.5 Hz, 1H), 4.08-3.85 (m, 1H), 3.44-3.06 (m, 2H), 2.97 (s, 3H), 2.15 (d, J=34.6 Hz, 2H), 1.50 (d, J=6.6 Hz, 3H). ES/MS 477.1 (M+H+).
  • (S)-2,4-diamino-6-(((5-chloro-3-(2-cyanoethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 112). 1H NMR (400 MHz, DMSO-d6) δ 7.77 (t, J=8.0 Hz, 1H), 7.64-7.56 (m, 2H), 5.09 (t, J=8.2 Hz, 1H), 4.37-4.09 (m, 2H), 3.14-2.98 (m, 1H), 2.97-2.80 (m, 1H), 2.95-2.79 (m, 1H), 1.74-1.56 (m, 1H), 0.64-0.52 (m, 2H), 0.51-0.41 (m, 2H). ES/MS 436.1 (M+1+).
  • (S)-2,4-diamino-6-(((5-chloro-3-(3-cyanopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 113). 1H NMR (400 MHz, DMSO-d6) δ 7.74 (dd, J=8.2, 7.8 Hz, 1H), 7.59 (dd, J=8.2, 1.2 Hz, 1H), 7.55 (dd, J=7.8, 1.2 Hz, 1H), 6.76-6.67 (m, 3H), 6.46 (br s, 3H), 5.16 (t, J=7.9 Hz, 1H), 4.21-4.11 (m, 1H), 4.07-3.97 (m, 1H), 2.64-2.57 (m, 2H), 2.06-1.95 (m, 2H), 1.66-1.56 (m, 1H), 0.59-0.52 (m, 2H), 0.46-0.40 (m, 2H). ES/MS 450.1 (M+1+).
  • (S)-2,4-diamino-6-(((5-chloro-3-(4-cyanobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 114). 1H NMR (400 MHz, DMSO-d6) δ 7.74 (dd, J=8.2, 7.8 Hz, 1H), 7.60 (dd, J=8.2, 1.2 Hz, 1H), 7.55 (dd, J=7.8, 1.2 Hz, 1H), 6.88 (d, J=8.2 Hz, 1H), 6.70 (br s, 2H), 6.47 (br s, 2H), 5.15 (t, J=8.0 Hz, 1H), 4.04-3.92 (m, 2H), 1.81-1.49 (m, 5H), 0.59-0.46 (m, 3H), 0.41-0.31 (m, 1H). ES/MS 464.2 (M+1+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-cyanopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)amino)pyrimidine-5-carbonitrile (Compound 115). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (dd, J=8.3, 7.7 Hz, 1H), 7.55 (dd, J=8.1, 1.1 Hz, 1H), 7.53 (dd, J=7.7, 1.2 Hz, 1H), 6.76 (d, J=7.8 Hz, 1H), 6.67 (br s, 2H), 5.34-5.15 (m, 1H), 4.43-3.87 (m, 2H), 2.26-1.69 (m, 6H), 0.99 (t, J=7.5 Hz, 3H). ES/MS 438.1 (M+1+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(4-cyanobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)amino)pyrimidine-5-carbonitrile (Compound 116). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (dd, J=8.2, 7.8 Hz, 1H), 7.56 (dd, J=8.2, 1.2 Hz, 1H), 7.54 (dd, J=7.8, 1.2 Hz, 1H), 6.84 (d, J=8.3 Hz, 1H), 6.67 (br s, 2H), 5.35-5.26 (m, 1H), 4.18-3.95 (m, 2H), 2.61-2.54 (m, 2H), 2.13-1.56 (m, 6H), 0.96 (t, J=7.2 Hz, 3H). ES/MS 452.2 (M+1+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)amino)pyrimidine-5-carbonitrile (Compound 117). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (dd, J=8.3, 7.7 Hz, 1H), 7.54 (dd, J=8.3, 1.2 Hz, 1H), 7.54 (dd, J=7.7, 1.2 Hz, 1H), 6.91 (br s, 2H), 5.33-5.23 (m, 1H), 4.43-4.32 (m, 1H), 4.05-3.94 (m, 1H), 3.41-3.26 (m, 2H), 3.02 (s, 3H), 2.40-2.22 (m, 1H), 2.22-2.06 (m, 1H), 1.98-1.84 (m, 2H), 1.01 (t, J=7.3 Hz, 3H). ES/MS 491.1 (M+1+).
  • (S)-2,4-diamino-6-(((5-chloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 118). 1H NMR (400 MHz, DMSO-d6) δ 7.73 (dd, J=8.2, 7.7 Hz, 1H), 7.57 (dd, J=8.2, 1.2 Hz, 1H), 7.55 (dd, J=7.8, 1.2 Hz, 1H), 6.73 (br s, 2H), 6.53 (d, J=8.3 Hz, 1H), 5.26 (t, J=7.9 Hz, 1H), 4.32-4.21 (m, 1H), 4.12-3.99 (m, 1H), 3.33 (s, 3H), 3.32-3.20 (m, 2H), 2.31-2.20 (m, 1H), 2.19-2.04 (m, 1H), 1.63-1.51 (m, 1H), 0.60-0.52 (m, 2H), 0.51-0.43 (m, 1H), 0.41-0.32 (m, 1H). ES/MS 503.1 (M+1+).
  • (S)-2,4-diamino-6-((cyclopropyl(5,8-dichloro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)methyl)amino)pyrimidine-5-carbonitrile (Compound 119). 1H NMR (400 MHz, DMSO-d6) δ 7.92 (d, J=8.5 Hz, 1H), 7.53 (d, J=8.5 Hz, 1H), 6.75 (br s, 2H), 6.42 (d, J=8.7 Hz, 1H), 5.42 (dd, J=8.7, 7.2 Hz, 1H), 4.33-4.22 (m, 1H), 4.10-3.99 (m, 1H), 3.32-3.21 (m, 2H), 2.99 (s, 3H), 2.31-2.06 (m, 2H), 1.62-1.51 (m, 1H), 0.65-0.43 (m, 3H), 0.43-0.30 (m, 1H). ES/MS 537.1 (M+1+).
  • (S)-2,4-diamino-6-(((8-chloro-6-fluoro-3-(3-(methylsulfonyl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 120). 1H NMR (400 MHz, DMSO-d6) δ 8.08 (dd, J=8.5, 2.9 Hz, 1H), 7.81 (dd, J=8.3, 2.9 Hz, 1H), 6.75 (br s, 2H), 6.46 (d, J=8.7 Hz, 1H), 5.42 (dd, J=8.5, 7.3 Hz, 1H), 4.38-4.24 (m, 1H), 4.17-4.04 (m, 1H), 3.32-3.24 (m, 2H), 2.31-2.08 (m, 2H), 1.65-1.53 (m, 1H), 0.61-0.43 (m, 3H), 0.40-0.32 (m, 1H). ES/MS 521.1 (M+1+).
  • (S)-4-(8-chloro-2-(cyclopropyl((2,6-diamino-5-cyanopyrimidin-4-yl)amino)methyl)-6-fluoro-4-oxoquinazolin-3(4H)-yl)butanamide (Compound 121). 1H NMR (400 MHz, DMSO-d6) δ 8.06 (dd, J=8.5, 2.9 Hz, 1H), 7.79 (dd, J=8.3, 2.9 Hz, 1H), 7.43 (s, 1H), 6.92 (s, 1H), 6.73 (s, 2H), 6.57 (s, 2H), 6.29 (d, J=8.7 Hz, 1H), 5.52-5.45 (m, 1H), 4.35-4.17 (m, 1H), 4.07-3.89 (m, 1H), 2.30-1.74 (m, 4H), 1.62-1.48 (m, 1H), 0.63-0.26 (m, 4H). ES/MS 486.1 (M+1+).
  • (S)-2,4-diamino-6-(((5-chloro-3-(4-(methylsulfonyl)butyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 122). 1H NMR (400 MHz, DMSO-d6) δ 7.74 (dd, J=8.2, 7.8 Hz, 1H), 7.60 (dd, J=8.2, 1.2 Hz, 1H), 7.55 (dd, J=7.8, 1.2 Hz, 1H), 6.86 (d, J=8.2 Hz, 1H), 6.71 (br s, 2H), 5.16 (t, J=8.0 Hz, 1H), 4.04-3.94 (m, 2H), 3.19-3.09 (m, 2H), 2.95 (s, 3H), 1.85-1.59 (m, 5H), 0.60-0.43 (m, 3H), 0.43-0.31 (m, 1H). ES/MS 517.1 (M+1+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-morpholinopropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 127). 1H NMR (400 MHz, DMSO-d6) δ 9.85 (s, 1H), 7.75 (q, J=8.0 Hz, 1H), 7.65-7.50 (m, 2H), 7.43 (s, 2H), 7.23-6.98 (s, 2H), 5.40 (p, J=6.6 Hz, 1H), 4.14-4.02 (m, 2H), 3.99 (d, J=13.0 Hz, 2H), 3.64 (t, J=12.3 Hz, 2H), 3.42 (s, 2H), 3.22 (d, J=10.1 Hz, 2H), 3.08 (s, 2H), 2.12 (p, J=7.8 Hz, 2H), 1.54 (d, J=6.5 Hz, 3H). ES/MS 484.2 (M+H+).
  • (S)-3-(5-chloro-2-(1-((2,6-diamino-5-cyanopyrimidin-4-yl)amino)ethyl)-4-oxoquinazolin-3(4H)-yl)-N,N-dimethylpropanamide (Compound 128). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (dd, J=8.2, 7.8 Hz, 1H), 7.61-7.48 (m, 2H), 7.07 (d, J=7.2 Hz, 1H), 6.63 (s, 2H), 6.44 (s, 2H), 5.41 (p, J=6.7 Hz, 1H), 4.40 (ddd, J=14.1, 9.7, 6.1 Hz, 1H), 4.04-3.89 (m, 1H), 2.95 (s, 3H), 2.89-2.81 (m, 1H), 2.79 (s, 3H), 1.51 (d, J=6.5 Hz, 3H). ES/MS 456.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(3-methoxy-3-methylbutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 129). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (dd, J=8.2, 7.8 Hz, 1H), 7.61-7.48 (m, 2H), 7.14 (d, J=7.4 Hz, 1H), 6.64 (s, 2H), 6.35 (s, 2H), 5.46-5.34 (m, 1H), 4.08 (dd, J=12.1, 6.4 Hz, 1H), 3.97-3.82 (m, 1H), 3.15 (s, 3H), 1.87 (tt, J=12.9, 6.2 Hz, 2H), 1.52 (d, J=6.5 Hz, 3H), 1.13 (d, J=8.1 Hz, 7H). ES/MS 456.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(2-ethoxyethyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 130). 1H NMR (400 MHz, DMSO-d6) δ 7.74 (dd, J=8.2, 7.8 Hz, 1H), 7.56 (ddd, J=10.8, 8.0, 1.2 Hz, 2H), 6.91 (d, J=7.3 Hz, 1H), 6.65 (s, 2H), 6.45 (s, 2H), 5.52 (t, J=6.8 Hz, 1H), 4.52-4.39 (m, 1H), 4.19-4.07 (m, 1H), 3.76-3.55 (m, 2H), 3.41 (dd, J=7.0, 3.9 Hz, 1H), 1.50 (d, J=6.5 Hz, 3H), 1.01 (t, J=7.0 Hz, 3H). ES/MS 429.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-3-(1,3-dimethoxypropan-2-yl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 131). 1H NMR (400 MHz, DMSO-d6) δ 7.76 (dd, J=8.2, 7.8 Hz, 1H), 7.65-7.50 (m, 2H), 7.17 (d, J=7.3 Hz, 1H), 6.63 (s, 2H), 6.51 (s, 2H), 5.50 (p, J=6.5 Hz, 1H), 4.44 (s, 1H), 4.08-3.95 (m, 1H), 3.91 (dd, J=9.7, 6.6 Hz, 1H), 3.72 (ddd, J=22.3, 9.9, 4.9 Hz, 2H), 3.24 (s, 3H), 3.17 (s, 3H), 1.99 (s, OH), 1.51 (d, J=6.3 Hz, 3H). ES/MS 459.2 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5-chloro-3-((S)-1-methoxypropan-2-yl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 132). 1H NMR (400 MHz, DMSO-d6) δ 7.79-7.68 (m, 1H), 7.60-7.50 (m, 2H), 7.18 (d, J=7.0 Hz, 1H), 6.71-6.59 (m, 2H), 6.49 (d, J=13.0 Hz, 2H), 5.45 (p, J=6.5 Hz, 1H), 4.45 (d, J=10.9 Hz, 1H), 3.93-3.76 (m, 2H), 3.22 (s, OH), 3.17 (s, 3H), 1.54 (d, J=6.7 Hz, 3H), 1.49 (d, J=6.5 Hz, 3H), 1.45 (d, J=6.7 Hz, 1H). ES/MS 429.2 (M+H+).
  • (S)-4-amino-6-((1-(5-chloro-3-(3-(4-methylpiperazin-1-yl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 133). 1H NMR (400 MHz, DMSO-d6) δ 8.1 (d, J=1.2 Hz, 1H), 7.92 (d, J=7.0 Hz, 1H), 7.75 (td, J=8.0, 1.2 Hz, 1H), 7.61 (ddt, J=7.5, 5.2, 1.2 Hz, 2H), 7.47 (d, J=24.2 Hz, 2H), 5.49 (p, J=6.5 Hz, 1H), 4.26-4.10 (m, 1H), 4.01-3.83 (m, 1H), 3.8-3.32 (m, 6H), 3.31-2.96 (m, 2H), 2.86 (s, 3H), 1.59 (dd, J=6.5, 1.3 Hz, 3H). ES/MS 468.2 (M+H+).
  • (S)-4-amino-6-((1-(5-chloro-4-oxo-3-(3-(piperidin-1-yl)propyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 134). 1H NMR (400 MHz, DMSO-d6) δ 9.12 (s, 1H), 8.05 (d, J=1.8 Hz, 1H), 7.83 (dd, J=6.7, 1.8 Hz, 1H), 7.74 (td, J=8.0, 1.9 Hz, 1H), 7.56 (dddd, J=7.7, 5.4, 2.1, 1.2 Hz, 2H), 7.44 (s, 1H), 5.42 (td, J=6.8, 2.1 Hz, 1H), 4.19 (dt, J=14.5, 6.5 Hz, 1H), 3.96 (ddd, J=14.3, 8.7, 5.6 Hz, 1H), 3.43 (t, J=10.2 Hz, 2H), 3.28-3.05 (m, 2H), 2.87 (p, J=10.1 Hz, 2H), 2.19 (ddt, J=32.8, 16.5, 8.0 Hz, 2H), 1.82 (d, J=14.3 Hz, 2H), 1.65 (dd, J=26.6, 13.8 Hz, 2H), 1.55 (dd, J=6.6, 1.9 Hz, 3H), 1.39 (t, J=12.5 Hz, 1H), 1.31-1.17 (m, 1H). ES/MS 467.2 (M+H+).
  • (S)-4-amino-6-((1-(5-chloro-3-(3-(4-methylpiperazin-1-yl)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 135). 1H NMR (400 MHz, DMSO-d6) δ 8.05 (d, J=1.2 Hz, 1H), 7.81 (d, J=7.0 Hz, 1H), 7.73 (td, J=8.0, 1.2 Hz, 1H), 7.55 (ddt, J=7.5, 5.2, 1.2 Hz, 2H), 7.40 (d, J=24.2 Hz, 2H), 5.45 (p, J=6.5 Hz, 1H), 4.26-4.10 (m, 1H), 4.01-3.83 (m, 1H), 3.66-2.96 (m, 6H), 2.80 (s, 3H), 2.04 (dd, J=19.8, 11.2 Hz, 2H), 1.55 (dd, J=6.5, 1.3 Hz, 3H), 1.32-1.19 (m, 1H). ES/MS 482.2 (M+H+).
  • (S)-4-amino-6-((1-(5-chloro-4-oxo-3-(2-(pyrrolidin-1-yl)ethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 136). 1H NMR (400 MHz, DMSO-d6) δ 9.12 (s, 1H), 8.05 (d, J=1.8 Hz, 1H), 7.74 (td, J=8.0, 1.9 Hz, 1H), 7.56 (dddd, J=7.7, 5.4, 2.1, 1.2 Hz, 2H), 7.44 (s, 2H), 5.42 (td, J=6.8, 2.1 Hz, 1H), 4.35 (qt, J=14.5, 6.8 Hz, 2H), 3.67 (d, J=11.7 Hz, 2H), 3.60-3.49 (m, 2H), 3.14 (dp, J=23.8, 7.9 Hz, 2H), 2.04 (s, 2H), 1.89 (q, J=11.4, 9.4 Hz, 2H), 1.53 (d, J=6.5 Hz, 3H). ES/MS 439.2 (M+H+).
  • (S)-2,4-diamino-6-((1-(5-chloro-4-oxo-3-(2-(pyrrolidin-1-yl)ethyl)-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 137). 1H NMR (400 MHz, DMSO-d6) δ 9.78 (s, 1H), 7.83-7.72 (m, 1H), 7.64-7.54 (m, 2H), 7.38-7.20 (m, 2H), 6.98 (s, 2H), 5.30 (p, J=6.6 Hz, 1H), 4.35 (qt, J=14.5, 6.8 Hz, 2H), 3.67 (d, J=11.7 Hz, 2H), 3.60-3.49 (m, 2H), 3.14 (dp, J=23.8, 7.9 Hz, 2H), 2.04 (s, 2H), 1.89 (q, J=11.4, 9.4 Hz, 2H), 1.53 (d, J=6.5 Hz, 3H). ES/MS 454.0 (M+H+).
  • (S)-4-((1-(3-(2-(1H-pyrazol-5-yl)ethyl)-5-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)-2,6-diaminopyrimidine-5-carbonitrile (Compound 140). 1H NMR (400 MHz, DMSO-d6) δ 7.80-7.70 (m, 1H), 7.63-7.51 (m, 4H), 6.09 (d, J=2.1 Hz, 1H), 5.38 (t, J=6.9 Hz, 1H), 4.59 (m, 1H), 4.39-4.27 (m, 2H), 4.06 (dd, J=14.5, 7.9 Hz, 2H), 3.05 (q, J=6.6, 5.1 Hz, 2H), 1.47 (d, J=6.5 Hz, 2H), 1.26 (d, J=5.9 Hz, 2H). ES/MS 451.1 (M+H+).

O. Preparation of a compound of formula (I) wherein n is 1, R1 is chloro, m is 0, B is (CH2)2, R2 is CONH2, R3 is cyclopropyl, R4 is 2,6-diamino-5-chloropyrimidin-4-yl, and R5 is hydrogen

To a mixture of (S)-3-(5-chloro-2-(cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)propanenitrile (46 mg, 0.10 mmol) in EtOH/H2O (1:1, 2 mL) was added hydrido(dimethylphosphinous acid-kP)[hydrogen bis(dimethylphosphinito-kP)]platinum (II) (6 mg, 0.01 mmol). The mixture stirred at 90° C. for 1 h. The mixture was cooled to room temperature and diluted with DCM/H2O and the aqueous layer was extracted with DCM. The combined organic layers were washed with brine, dried (Na2SO4), filtered and concentrated. The residue was purified via flash chromatography (SiO2, 0-50% MeOH/EtOAc) to give (S)-3-(5-chloro-2-(cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)propanamide (Compound 68). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (t, J=8.1 Hz, 1H), 7.56 (dd, J=8.2, 1.2 Hz, 1H), 7.54 (dd, J=7.8, 1.2 Hz, 1H), 7.49 (br s, 1H), 6.99 (br s, 1H), 6.18-6.00 (m, 3H), 5.66 (br s, 2H), 5.23 (t, J=7.8 Hz, 1H), 4.58-4.37 (m, 1H), 4.24-4.07 (m, 1H), 2.78-2.63 (m, 1H), 2.63-2.53 (m, 1H), 1.63-1.47 (m, 1H), 0.59-0.42 (m, 3H), 0.39-0.30 (m, 1H). ES/MS 463.1 (M+H+).

P. Preparation of the below compounds of Formula (I), using Example 30 and Reaction Scheme I:

  • (S)-4-(5-chloro-2-(cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-4-oxoquinazolin-3(4H)-yl)butanamide (Compound 69). 1H NMR (400 MHz, DMSO-d6) δ 7.70 (t, J=8.1 Hz, 1H), 7.54 (dd, J=8.2, 1.2 Hz, 1H), 7.52 (dd, J=7.8, 1.2 Hz, 1H), 7.43 (br s, 1H), 6.90 (br s, 1H), 6.10 (s, 2H), 5.90 (d, J=8.9 Hz, 1H), 5.79 (s, 2H), 5.30 (dd, J=8.9, 7.3 Hz, 1H), 4.33-4.23 (m, 1H), 3.97-3.87 (m, 1H), 2.35-2.18 (m, 3H), 1.91-1.78 (m, 1H), 1.56-1.46 (m, 1H), 0.57-0.47 (m, 3H), 0.34-0.27 (m, 1H). ES/MS 477.1 (M+H+).

Q. Preparation of a compound of formula (I) wherein n is 1, R1 is chloro, m is 0, B is cyclobutyl, R2 is OH, R3 is methyl, R4 is 2,6-diamino-5-cyanopyrimidin-4-yl, and R5 is hydrogen

A solution of (1R,3s)-3-(2-((S)-1-((6-amino-5-cyanopyrimidin-4-yl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)cyclobutyl benzoate (27 mg, 0.052 mmol) dissolved in tetrahydrofuran (700 μL) and methanol (300 μL) was treated with lithium hydroxide (7 mg, 0.16 mmol) dissolved in water. The reaction mixture was stirred at room temperature for 1 h and concentrated in vacuo. The residue was suspended in EtOAc and washed with 1N HCl solution. The organic layer was washed with brine, dried over Na2SO4, and concentrated to give a light-yellow solid. The solid was dissolved in DMF and purified by HPLC to give 4-amino-6-(((S)-1-(5-chloro-3-((1S,3R)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 82). 1H NMR (400 MHz, DMSO-d6) δ 8.07 (s, 1H), 7.76 (m, 1H), 7.69 (m, 1H), 7.50 (d, J=8 Hz, 2H), 7.39 (m, 1H), 5.55 (m, 1H), 4.12 (m, 1H), 3.73 (m, 1H), 2.80 (m, 2H), 2.57 (m, 1H), 2.47 (m, 1H), 1.46 (d, J=6.4 Hz, 3H). ES/MS 412.0 (M+H+).

R. Preparation of the below compounds of formula (I), using Example 3Q and Reaction Scheme I:

  • (S)-4-amino-6-((1-(5-chloro-3-(3-hydroxypropyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 83). 1H NMR (400 MHz, DMSO-d6) δ 8.00 (s, 1H), 7.70 (m, 1H), 7.52 (m, 1H), 7.34 (m, 1H), 5.46 (m, 1H), 4.21 (m, 2H), 3.92 (m, 2H), 1.88 (m, 2H), 1.52 (d, J=6.8 Hz, 3H). ES/MS 400.1 (M+H+).
  • 4-amino-6-(((1S)-1-(5-chloro-3-(4-hydroxybutan-2-yl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 84). 1H NMR (400 MHz, DMSO-d6) δ 8.04 (s, 1H), 7.69 (m, 1H), 7.59 (m, 1H), 7.52 (m, 2H), 5.65 (m, 1H), 4.40 (m, 1H), 3.38 (m, 1H), 3.15 (m, 1H), 2.24 (m, 1H), 1.96 (m, 1H), 1.52 (d, J=8 Hz, 3H), 1.47 (d, J=8 Hz, 3H). ES/MS 414.1 (M+H+).
  • (S)-4-amino-6-((1-(5-chloro-3-(4-hydroxycyclohexyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 85). 1H NMR (400 MHz, DMSO-d6) 8.03 (s, 1H), 7.71 (m, 1H), 7.67 (m, 1H), 7.49 (m, 1H), 5.70 (m, 1H), 3.85 (m, 1H), 1.73 (m, 1H), 1.70 (m, 1H), 1.62 (m, 1H), 1.50 (d, J=8 Hz, 3H). ES/MS 440.1 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5-chloro-3-((cis)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 93). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (dd, J=8.2, 7.8 Hz, 1H), 7.56-7.52 (m, 2H), 5.52 (p, J=6.7 Hz, 1H), 4.14 (p, J=8.2 Hz, 1H), 3.85-3.74 (m, 1H), 2.80-2.61 (m, 4H), 1.49 (d, J=6.5 Hz, 3H). ES/MS 427.1 (M+H+).
  • 5-chloro-2-((S)-1-((2,6-diamino-5-chloropyrimidin-4-yl)amino)ethyl)-3-((cis)-3-hydroxycyclobutyl)quinazolin-4(3H)-one (Compound 94). 1H NMR (400 MHz, DMSO-d6) δ 7.72 (t, J=8.2 Hz, 1H), 7.59-7.52 (m, 2H), 5.53-5.44 (m, 1H), 4.13 (p, J=8.2 Hz, 1H), 3.81 (p, J=7.0 Hz, 1H), 2.81-2.60 (m, 4H), 1.50 (d, J=6.5 Hz, 3H). ES/MS 436.1 (M+H+).
  • 5,8-dichloro-2-((S)-cyclopropyl((2,6-diamino-5-chloropyrimidin-4-yl)amino)methyl)-3-((cis)-3-hydroxycyclobutyl)quinazolin-4(3H)-one (Compound 95). 1H NMR (400 MHz, DMSO-d6) δ 7.93 (d, J=8.5 Hz, 1H), 7.55 (d, J=8.5 Hz, 1H), 5.27 (t, J=7.7 Hz, 1H), 4.21-4.09 (m, 1H), 3.81 (t, J=7.1 Hz, 1H), 2.84-2.63 (m, 4H), 1.61-1.47 (m, 1H), 0.64-0.39 (m, 4H). ES/MS 496.1 (M+H+).
  • 2,4-diamino-6-(((S)-cyclopropyl(5,8-dichloro-3-((cis)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)methyl)amino)pyrimidine-5-carbonitrile (Compound 96). 1H NMR (400 MHz, DMSO-d6) δ 7.89 (d, J=8.5 Hz, 1H), 7.51 (d, J=8.3 Hz, 1H), 5.27 (t, J=7.7 Hz, 1H), 4.12 (t, J=8.3 Hz, 1H), 3.76 (p, J=7.3 Hz, 1H), 2.80-2.57 (m, 4H), 1.52 (dt, J=8.0, 6.4 Hz, 1H), 0.62-0.32 (m, 4H). ES/MS 487.1 (M+H+).
  • 2,4-diamino-6-(((S)-(8-chloro-6-fluoro-3-((trans)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 97). 1H NMR (400 MHz, DMSO-d6) δ 8.04 (dd, J=8.5, 2.9 Hz, 1H), 7.79 (dd, J=8.3, 2.9 Hz, 1H), 5.38 (t, J=7.2 Hz, 1H), 5.03-4.95 (m, 1H), 4.55-4.48 (m, 2H), 3.23-3.03 (m, 2H), 2.21-1.98 (m, 2H), 1.51-1.43 (m, 1H), 0.60-0.31 (m, 4H). ES/MS 471.1 (M+H+).
  • 2,4-diamino-6-(((S)-1-(8-chloro-6-fluoro-3-((trans)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 98). 1H NMR (400 MHz, DMSO-d6) δ 8.05 (dd, J=8.5, 2.9 Hz, 1H), 7.80 (dd, J=8.3, 2.9 Hz, 1H), 5.57 (t, J=6.7 Hz, 1H), 4.92-4.82 (m, 1H), 4.54-4.47 (m, 1H), 3.21-3.05 (m, 2H), 2.20-2.09 (m, 1H), 2.09-2.00 (m, 1H), 1.48 (d, J=6.4 Hz, 3H). ES/MS 445.1 (M+H+).
  • 2,4-diamino-6-(((S)-1-(8-chloro-6-fluoro-3-((cis)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 99). 1H NMR (400 MHz, DMSO-d6) δ 8.03 (dd, J=8.5, 2.9 Hz, 1H), 7.77 (dd, J=8.3, 2.9 Hz, 1H), 5.59 (p, J=7.1, 6.6 Hz, 1H), 4.14 (p, J=8.4, Hz, 1H), 3.76 (p, J=7.1 Hz, 1H), 2.94-2.77 (m, 2H), 2.61-2.51 (m, 1H), 2.45-2.39 (m, 1H), 1.48 (d, J=6.4 Hz, 3H). ES/MS 445.1 (M+H+).
  • 2,4-diamino-6-(((S)-cyclopropyl(5,8-dichloro-3-((trans)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)methyl)amino)pyrimidine-5-carbonitrile (Compound 100). 1H NMR (400 MHz, DMSO-d6) δ 7.89 (d, J=8.5 Hz, 1H), 7.51 (d, J=8.8 Hz, 1H), 5.35 (t, J=7.4 Hz, 1H), 4.99-4.89 (m, 1H), 4.53-4.45 (m, 1H), 3.16-2.99 (m, 2H), 2.21-2.11 (m, 1H), 2.09-1.98 (m, 1H), 1.52-1.41 (m, 1H), 0.60-0.31 (m, 4H). ES/MS 487.1 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5,8-dichloro-3-((trans)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 101). 1H NMR (400 MHz, DMSO-d6) δ 7.89 (dd, J=8.5, 0.7 Hz, 1H), 7.51 (dd, J=8.5, 0.9 Hz, 1H), 5.52 (t, J=7.0 Hz, 1H), 4.89-4.80 (m, 1H), 4.52-4.44 (m, 1H), 3.14-3.01 (m, 2H), 2.19-2.10 (m, 1H), 2.10-2.01 (m, 1H), 1.46 (d, J=6.3 Hz, 3H). ES/MS 461.1 (M+H+).
  • 2,4-diamino-6-(((S)-(8-chloro-6-fluoro-3-((cis)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)(cyclopropyl)methyl)amino)pyrimidine-5-carbonitrile (Compound 102). 1H NMR (400 MHz, DMSO-d6) δ 8.03 (dd, J=8.7, 2.9 Hz, 1H), 7.77 (dd, J=8.3, 2.5 Hz, 1H), 5.30 (t, J=7.8 Hz, 1H), 4.18 (p, J=8.6 Hz, 1H), 3.79-3.69 (m, 1H), 2.92 (q, J=8.7 Hz, 1H), 2.83 (q, J=9.2 Hz, 1H), 2.60-2.51 (m, 1H), 2.41-2.31 (m, 1H), 1.58-1.49 (m, 1H), 0.60-0.31 (m, 4H). ES/MS 471.1 (M+H+).
  • 2,4-diamino-6-(((S)-1-(5,8-dichloro-3-((cis)-3-hydroxycyclobutyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile (Compound 103). 1H NMR (400 MHz, DMSO-d6) δ 7.88 (d, J=8.5 Hz, 1H), 7.50 (d, J=8.5 Hz, 1H), 5.52 (p, J=6.8 Hz, 1H), 4.08 (p, J=8.3 Hz, 1H), 3.82-3.73 (m, 1H), 2.76-2.56 (m, 4H), 1.47 (d, J=6.4 Hz, 3H). ES/MS 461.1 (M+H+).

Example 4 Preparation of a Compound of Formula (4)

A. Preparation of a compound of formula (4) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, R2y is methyl, R3 is methyl, and R5 is hydrogen


(S)-(9H-fluoren-9-yl)methyl (1-(5-chloro-3-(3-(methylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate was prepared as described above from (S)-tert-butyl (3-(2-(1-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethyl)-5-chloro-4-oxoquinazolin-3(4H)-yl)propyl)(methyl)carbamate.

Example 5 Preparation of a Compound of Formula (5)

A. Preparation of a compound of formula (5) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, R2y is methyl, R2x is trifluoroethyl, R3 is methyl, and R5 is hydrogen

To a mixture of (S)-(9H-fluoren-9-yl)methyl (1-(5-chloro-3-(3-(methylamino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate (0.21 g, 0.40 mmol) and potassium carbonate (0.33 g, 2.4 mmol) in MeCN (8 mL) was added 2,2,2-trifluoroethyltriflate (70 μL, 0.49 mmol). The mixture was heated to 55° C. overnight. The mixture was filtered and the filtrate was concentrated. The residue was partitioned between EtOAc and H2O and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with brine, dried (Na2SO4), decanted and concentrated. The residue was carried forward without purification to give (S)-(9H-fluoren-9-yl)methyl (1-(5-chloro-3-(3-(methyl(2,2,2-trifluoroethyl)amino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate.

Example 6 Preparation of a Compound of Formula (6)

A. Preparation of a Compound of Formula (6) wherein n is 1, R1 is chloro, m is 0, B is (CH2)3, R2y is methyl, R2x is trifluoroethyl, R3 is methyl, and R5 is hydrogen

(S)-2-(1-aminoethyl)-5-chloro-3-(3-(methyl(2,2,2-trifluoroethyl)amino)propyl)quinazolin-4(3H)-one was prepared as described above from (S)-(9H-fluoren-9-yl)methyl (1-(5-chloro-3-(3-(methyl(2,2,2-trifluoroethyl)amino)propyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)carbamate.

Biological Examples

The compounds of formula (I) were characterized for their enzymatic activity against the PI3K isoforms. The activities were measured using a time-resolved fluorescence resonance energy transfer (TR-FRET) assay. TR-FRET monitored the formation of 3,4,5-inositol triphosphate molecule that competed with fluorescently labeled PIP3 for binding to the GRP-1 pleckstrin homology domain protein. An increase in phosphatidylinositide 3-phosphate product resulted in a decrease in TR-FRET signal as the labeled fluorophore was displaced from the GRP-1 protein binding site.

Class I PI3K isoforms were expressed and purified as heterodimeric recombinant proteins. All assay reagents and buffers for the TR-FRET assay were purchased from Millipore. PI3K isoforms were assayed under initial rate conditions in the presence of 25 mM Hepes (pH 7.4), and 2×Km ATP (75-500 μM), 2 μM PIP2, 5% glycerol, 5 mM MgCl2, 50 mM NaCl, 0.05% (v/v) Chaps, 1 mM dithiothreitol, and 1% (v/v) DMSO at the following concentrations for each isoform: PI3Kα, PI3Kβ, and PI3Kδ between 25 and 50 pM, and PI3Kγ at 2 nM. The compounds of Table 1 and Compound X, ((S)-2,4-diamino-6-((1-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile) were added to the assay solution and incubated for 30 minutes at 25° C. The reactions were terminated with a final concentration of 10 mM EDTA, 10 nM labeled-PIP3, and 35 nM Europium labeled GRP-1 detector protein before reading TR-FRET on an Envision plate reader (Ex: 340 nm; Em: 615/665 nm; 100 μs delay and 500 μs read window).

The results were normalized based on positive (1 pM wortmanin) and negative (DMSO) controls, and the IC50 values for PI3K α, β, δ, and γ were calculated from the fit of the dose-response curves to a four-parameter equation. These assays generally produced results within 3-fold of the reported mean.

Table 2 summarizes the IC50 (nM) values for PI3K isoforms β, δ, and γ. The results indicate that certain compounds of formula (I) inhibit both PI3Kδ and PI3Kβ. Also, Compound X exhibited PI3Kδ IC50 of 0.2 nM, a PI3Kβ IC50 of 11 nM, and a PI3Kγ IC50 of 7 nM. The PI3Kγ/PI3Kβ ratio for Compound X and compound 47 is 0.6 and 230, respectively. The results indicate that certain compounds have greater selectivity for PI3Kβ over PI3Kγ compared to compound X. Compounds in Table 1a were analyzed using the same assay, and the results are summarized in Table 2a.

TABLE 2 The IC50 values (nM) for PI3K isoforms β, δ, and γ. Compound IC50 (β) IC50 (δ) IC50 (γ) 1 27 9 ≥10000 2 1500 27 ≥10000 3 1300 15 ≥10000 4 420 5 660 5 930 5 2200 6 250 9 ≥10000 7 140 35 ≥10000 8 68 5 1700 9 170 77 ≥10000 10 840 480 ≥10000 11 93 37 ≥10000 12 300 52 ≥10000 13 6800 3300 ≥10000 14 9 2 1900 15 2 3 1700 16 42 7 ≥10000 17 69 11 ≥10000 18 360 370 ≥10000 19 1300 14 ≥10000 23 90 25 ≥10000 24 51 6 ≥10000 25 1300 100 ≥10000 26 740 150 ≥10000 27 200 7 ≥10000 28 100 3 ≥10000 29 47 7 ≥10000 30 110 11 ≥10000 31 47 6 ≥10000 32 44 92 3200 33 2100 210 ≥10000 34 250 79 ≥10000 35 110 21 ≥10000 36 71 27 8200 37 57 16 4800 38 44 12 ≥10000 39 25 3 2400 40 21 9 ≥10000 41 15 3 1800 42 15 2 1400 43 7 6 3400 44 8 11 6000 45 7 4 2100 46 6 3 2500 47 5 3 1100 48 4 4 900 49 1 1 750 50 75 71 4600 52 29 4 1200 51 110 8 5500 53 9 2 190 54 5 0.4 830 55 4 0.5 280 56 4 0.4 540 57 480 6 ≥10000 58 460 24 6600 59 500 8 2200 60 150 4 6400 61 140 7 6600 62 320 4 960 63 640 2 800 64 12 1 110 65 7 1 470 66 4 1 97 68 520 5 1000 69 49 2 1300 71 240 34 ≥10000 72 35 12 7900 73 21 6 9400 74 17 1 1620 75 19 1 2300

TABLE 2a The IC50 values (nM) for PI3K isoforms β, δ, and γ. Compound IC50 (β) IC50 (δ) IC50 (γ) 76 46 2 160 77 860 33 7000 78 920 49 3000 79 40 2 380 80 1100 27 2400 81 47 5 320 82 150 5 2300 83 180 6 2900 84 260 4 2900 85 92 3 1200 86 620 24 ≥10000 87 150 4 ≥10000 88 250 42 ≥10000 89 54 3 ≥10000 90 2800 37 ≥10000 91 1700 95 ≥10000 92 320 7 ≥10000 93 4 1 158 94 190 14 1587 95 720 16 ≥10000 96 10 2 960 97 58 8 2800 98 330 20 3200 99 290 9 2400 100 44 3 2900 101 140 12 3200 102 66 2 1100 103 21 5 2600 104 58 9 78 105 820 9 8400 106 590 8 >10000 107 82 3 1700 108 2000 39 ≥10000 109 520 14 2185 110 42 4 180 111 40 3 530 112 8 3 62 113 17 1 59 114 12 0.8 130 115 190 9 190 116 150 5 540 117 250 21 3000 118 13 2 570 119 8 2 1100 120 30 8 1900 121 9 2 760 122 23 0.7 120 123 79 3 1300 124 88 2 3200 125 49 3 2300 126 24 0.9 2500 127 130 2 720 128 530 15 280 129 290 2 530 130 120 3 410 131 210 13 1400 132 58 2 330 133 340 11 ≥10000 134 550 4 ≥10000 135 640 6 ≥10000 136 990 25 ≥10000 137 760 27 ≥10000 140 410 6 590 142 527 87 ≥10000 143 374 34 4236

All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification are incorporated herein by reference, in their entirety to the extent not inconsistent with the present description. From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the present application.

Claims

1. A compound having the structure of formula (IVa):

wherein:
n is 0, 1, 2, 3, or 4;
m is 0;
A is a single bond or C(O);
B is C1-6 alkyl or C3-8 cycloalkyl wherein each of the alkyl and cycloalkyl is optionally substituted with hydroxyl or C1-6 alkoxy;
each R1 is independently selected from halo, cyano, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted sulfonyl, optionally substituted C1-6 alkylsulfonyl, optionally substituted C3-8 aryl, optionally substituted C3-8 heteroaryl, optionally substituted C3-8 cycloalkyl, and optionally substituted C2-8 heterocycloalkyl;
R2 is cyano, optionally substituted C3-8 heterocycloalkyl, optionally substituted C3-8 heteroaryl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, or —SO2R2z, where R2x is independently hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, or C4-8 heteroaryl; wherein R2y is independently selected from hydrogen and C1-6 alkyl; and wherein R2z is C1-6 alkyl;
R3 is hydrogen, optionally substituted C1-6 alkyl, C3-8 cycloalkyl, or C6-10 aryl;
R4 is a six- to twelve-membered bicyclic heteroaryl having at least one aromatic group and at least two heteroatoms selected from N and S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted C1-6 alkyl, and —NH2; and
R5 is hydrogen or optionally substituted C1-6 alkyl;
or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof.

2. The compound of claim 1, wherein:

n is 0, 1, 2 or 3;
m is 0;
A is a single bond or C(O);
B is C1-6 alkyl or C3-8 cycloalkyl—wherein each of the alkyl and cycloalkyl is optionally substituted with hydroxyl or C1-6 alkoxy;
each R1 is independently selected from halo, cyano, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 alkylsulfonyl;
R2 is cyano, morpholinyl, —NR2xR2x, —NR2yC(O)R2x, —C(O)NR2xR2y, —OR2y, —SO2R2z, pyrrolidinyl, piperidinyl, optionally substituted piperazinyl, pyrazolyl, or triazolyl, where R2x is independently hydrogen, optionally substituted C1-6 alkyl, C1-6 haloalkyl, C6-10 aryl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-8 heterocycloalkyl, or C4-8 heteroaryl; wherein R2y is independently selected from hydrogen and C1-6 alkyl; and wherein R2z is C1-6 alkyl;
R3 is hydrogen, optionally substituted C1-6 alkyl, C3-8 cycloalkyl, or C6-10 aryl;
R4 is a six- to twelve-membered bicyclic heteroaryl having at least one aromatic group and at least two heteroatoms selected from N and S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted C1-6 alkyl, and —NH2; and
R5 is hydrogen or optionally substituted C1-6 alkyl;
or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof.

3. A compound selected from the group consisting of:

(S)-3-(3-amino-3-methylbutyl)-5-chloro-2-(1-(thiazolo[5,4-d]pyrimidin-7-ylamino)ethyl)quinazolin-4(3H)-one;
(S)-2-(1-((9H-purin-6-yl)amino)ethyl)-5-chloro-3-(3-morpholinopropyl)quinazolin-4(3H)-one;
(S)-3-(3-aminopropyl)-5-chloro-2-(cyclopropyl(imidazo[2,1-f][1,2,4]triazin-4-ylamino)methyl)quinazolin-4(3H)-one;
(S)-3-(3-aminopropyl)-2-(((5-bromo-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)(cyclopropyl)methyl)-5-chloroquinazolin-4(3H)-one;
(S)-3-(3-aminopropyl)-5-chloro-2-(cyclopropyl(thiazolo[5,4-d]pyrimidin-7-ylamino)methyl)quinazolin-4(3H)-one;
(S)-3-(3-aminopropyl)-5-chloro-2-(((6-chloropyrido[3,2-d]pyrimidin-4-yl)amino)(cyclopropyl)methyl)quinazolin-4(3H)-one;
(S)-3-(3-aminopropyl)-5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)quinazolin-4(3H)-one;
(S)-3-(3-aminopropyl)-5-chloro-2-(1-(thiazolo[5,4-d]pyrimidin-7-ylamino)ethyl)quinazolin-4(3H)-one; and
(S)-3-((1H-pyrazol-3-yl)methyl)-5-chloro-2-(1-(imidazo[2,1-f][1,2,4]triazin-4-ylamino)ethyl)quinazolin-4(3H)-one;
or a pharmaceutically acceptable salt thereof.

4. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, wherein B is methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with methoxy, ethoxy, or hydroxyl.

5. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, wherein each R1 is independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, and fluoroethyl.

6. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, wherein each R2 is selected from cyano, morpholinyl, —NH2, —NHR2x, —NR2xR2x, —NHC(O)R2y, —NR2yC(O)R2x, —C(O)NHR2y, —C(O)NR2xR2y, —OH, —OR2y, and —SO2R2z; wherein each R2x is independently methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, fluoromethyl, trifluoroethyl, difluoroethyl, fluoroethyl, phenyl, phenylmethyl, phenylethyl, pyridinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclohexyl, or oxetanyl, each of R2x is optionally substituted with one, two, or three members selected from fluoro, chloro, bromo, and iodo; wherein each R2y is independently hydrogen, methyl, ethyl, propyl, or butyl; and wherein each R2z is independently methyl, ethyl, propyl, or butyl.

7. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, wherein R3 is selected from hydrogen, methyl, ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutyl, cyclopentyl, and phenyl.

8. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, wherein R5 is hydrogen, methyl, ethyl, or propyl.

9. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, wherein R4 is bicyclic heteroaryl having at least one aromatic ring, at least two nitrogen atoms, and at least one additional heteroatom selected from N, O, and S; wherein R4 is optionally substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and —NH2.

10. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is an atropisomer.

11. A pharmaceutical composition comprising the compound of claim 1 or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, and at least one pharmaceutically acceptable vehicle.

12. A method of treating a disease or condition in a human in need thereof comprising administering to the human a therapeutically effective amount of the compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, wherein the disease or condition is selected from cancer, hematologic malignancies, leukemias, lymphomas, myeloproliferative disorders, myelodysplastic syndromes, plasma cell neoplasms, solid tumor, inflammation, fibrosis, autoimmune disorders, allergic conditions, hypersensitivity, cardiovascular diseases, neurodegenerative diseases, renal disorders, viral infections, obesity, and autoimmune diseases.

13. The method of claim 12, wherein the disease or condition is selected from rheumatoid arthritis, osteoarthritis, atherosclerosis, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, asthma, chronic obstructive airways disease, pneumonitis, dermatitis, alopecia, nephritis, vasculitis, atherosclerosis, Alzheimer's disease, hepatitis, primary biliary cirrhosis, sclerosing cholangitis, diabetes, acute rejection of transplanted organs, lymphomas, multiple myelomas, leukemias, pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, rectum cancer, liver cancer, kidney cancer, stomach cancer, skin cancer, gastric cancer, esophageal cancer, head and neck cancer, melanoma, neuroendocrine cancers, CNS cancers, brain tumors, bone cancer, and soft tissue sarcoma.

14. A method of inhibiting the activity of a phosphatidylinositol 3-kinase polypeptide by contacting the polypeptide with the compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof.

15. A method of inhibiting excessive or destructive immune reactions or growth or a proliferation of cancer cells, comprising administering an effective amount of the compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof.

16. A kit comprising the compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof, or a label and/or instructions for use.

17. The compound, or a pharmaceutically acceptable salt, stereoisomer, atropisomer, or a mixture thereof of claim 1 for use in therapy.

Referenced Cited
U.S. Patent Documents
3322756 May 1967 Ruschig et al.
3691016 September 1972 Patel
3897432 July 1975 Shen et al.
3969287 July 13, 1976 Jaworek et al.
3984555 October 5, 1976 Amschler et al.
4179337 December 18, 1979 Davis et al.
4183931 January 15, 1980 Wolfe et al.
4195128 March 25, 1980 Hidebrand et al.
4225489 September 30, 1980 Rolf et al.
4229537 October 21, 1980 Hodgins et al.
4247642 January 27, 1981 Hirohara et al.
4289872 September 15, 1981 Denkewalter et al.
4301144 November 17, 1981 Iwashita et al.
4330440 May 18, 1982 Ayers et al.
4496689 January 29, 1985 Mitra
4640835 February 3, 1987 Shimizu et al.
4670417 June 2, 1987 Iwasaki et al.
4791192 December 13, 1988 Nakagawa et al.
4925673 May 15, 1990 Steiner et al.
5013556 May 7, 1991 Woodle et al.
5225347 July 6, 1993 Goldberg et al.
5229490 July 20, 1993 Tam
5378725 January 3, 1995 Bonjouklian et al.
5480906 January 2, 1996 Creemer et al.
RE35862 July 28, 1998 Steiner et al.
5858753 January 12, 1999 Chantry et al.
5882910 March 16, 1999 Chantry et al.
5948664 September 7, 1999 Williams et al.
5985589 November 16, 1999 Chantry et al.
6046049 April 4, 2000 Monia et al.
6048970 April 11, 2000 Lal et al.
6277981 August 21, 2001 Tu et al.
6291220 September 18, 2001 Williams et al.
6369038 April 9, 2002 Blumenfeld et al.
6410224 June 25, 2002 Stinchcomb et al.
6426337 July 30, 2002 Cox et al.
6482623 November 19, 2002 Vanhaesebroeck et al.
6518277 February 11, 2003 Sadhu et al.
6667300 December 23, 2003 Sadhu et al.
6696250 February 24, 2004 Cech et al.
6800620 October 5, 2004 Sadhu et al.
6949535 September 27, 2005 Sadhu et al.
7932260 April 26, 2011 Fowler et al.
8138195 March 20, 2012 Sadhu et al.
8207153 June 26, 2012 Fowler et al.
8435988 May 7, 2013 Qu et al.
8492389 July 23, 2013 Sadhu et al.
RE44599 November 12, 2013 Fowler et al.
8586597 November 19, 2013 Fowler et al.
RE44638 December 10, 2013 Fowler et al.
8623881 January 7, 2014 Sadhu et al.
8637533 January 28, 2014 Sadhu et al.
8653077 February 18, 2014 Sadhu et al.
8673906 March 18, 2014 Barlaam et al.
8779131 July 15, 2014 Kesicki et al.
8980901 March 17, 2015 Fowler et al.
8993583 March 31, 2015 Fowler et al.
9018221 April 28, 2015 Evarts et al.
9029384 May 12, 2015 Evarts et al.
9149477 October 6, 2015 Kesicki et al.
9221795 December 29, 2015 Evarts et al.
9266878 February 23, 2016 Everts et al.
9340547 May 17, 2016 Anchuela et al.
9487772 November 8, 2016 Sadhu et al.
9499523 November 22, 2016 Kim
9676759 June 13, 2017 Aronov et al.
9765060 September 19, 2017 Kaplan et al.
10010550 July 3, 2018 Sadhu et al.
10092563 October 9, 2018 Cai et al.
10221197 March 5, 2019 Du et al.
10308639 June 4, 2019 Evarts et al.
10336756 July 2, 2019 Kesicki et al.
10398695 September 3, 2019 Sadhu et al.
20040023390 February 5, 2004 Davidson et al.
20040092561 May 13, 2004 Ruckle et al.
20040121996 June 24, 2004 Barvian et al.
20040138199 July 15, 2004 Goglietti et al.
20040242631 December 2, 2004 Garlich et al.
20040248953 December 9, 2004 Gogliotti et al.
20040248954 December 9, 2004 Gogliotti et al.
20040259926 December 23, 2004 Bruendl et al.
20040266780 December 30, 2004 Sadhu et al.
20050004195 January 6, 2005 Para et al.
20050020630 January 27, 2005 Connolly et al.
20050020631 January 27, 2005 Gogliotti et al.
20050043239 February 24, 2005 Douangpanya et al.
20050054614 March 10, 2005 Diacovo et al.
20050239809 October 27, 2005 Watts et al.
20060079538 April 13, 2006 Hallahan et al.
20060106038 May 18, 2006 Bouscary et al.
20080287469 November 20, 2008 Diacovo et al.
20090312319 December 17, 2009 Ren et al.
20100029693 February 4, 2010 Douangpanya et al.
20100202963 August 12, 2010 Gallatin et al.
20100249155 September 30, 2010 Evarts et al.
20100256167 October 7, 2010 Fowler et al.
20110044942 February 24, 2011 Puri et al.
20110230465 September 22, 2011 Stammers et al.
20120015964 January 19, 2012 Fowler et al.
20120040980 February 16, 2012 Huggins et al.
20130053362 February 28, 2013 Castro et al.
20130274253 October 17, 2013 Brollo et al.
20140154772 June 5, 2014 Sadhu et al.
20140179673 June 26, 2014 Evarts et al.
20140378479 December 25, 2014 Kesicki et al.
20150087663 March 26, 2015 Xi
20150361054 December 17, 2015 Cai et al.
20150361070 December 17, 2015 Evarts et al.
20150361095 December 17, 2015 Evarts et al.
20160075705 March 17, 2016 Kesicki et al.
20170049772 February 23, 2017 Sadhu et al.
20170340633 November 30, 2017 Sadhu et al.
20180065953 March 8, 2018 Evarts et al.
20180360832 December 20, 2018 Sadhu et al.
Foreign Patent Documents
2 895 782 June 2014 CA
1440408 September 2003 CN
101031569 September 2007 CN
103649089 March 2014 CN
0 525 960 February 1993 EP
0 525 960 February 1993 EP
0 675 124 October 1995 EP
0 675 124 October 1995 EP
0 716 857 June 1996 EP
0 716 857 June 1996 EP
0 884 310 December 1998 EP
0 884 310 December 1998 EP
0 900 568 March 1999 EP
0 900 568 March 1999 EP
1 356 763 June 1974 GB
1 554 057 October 1979 GB
2 017 097 October 1979 GB
55 118917 September 1980 JP
55 118918 January 1981 JP
56 002322 January 1981 JP
2003-512461 April 2003 JP
2005-509635 April 2005 JP
2007-537291 December 2007 JP
2013-515074 May 2013 JP
2013-526586 June 2013 JP
2013-540746 November 2013 JP
2016-503805 February 2016 JP
2016-527238 September 2016 JP
6125663 May 2017 JP
2017-517527 June 2017 JP
2017-517542 June 2017 JP
6207100 October 2017 JP
WO-93/21259 October 1993 WO
WO-94/17090 August 1994 WO
WO-95/24379 September 1995 WO
WO-96/04923 February 1996 WO
WO-96/25488 August 1996 WO
WO-96/32478 October 1996 WO
WO-97/34631 September 1997 WO
WO-97/41097 November 1997 WO
WO-97/43276 November 1997 WO
WO-97/46688 December 1997 WO
WO-98/33802 August 1998 WO
WO-98/38173 September 1998 WO
WO-99/08501 February 1999 WO
WO-99/08501 February 1999 WO
WO-99/34804 July 1999 WO
WO-01/00881 January 2001 WO
WO-01/30768 May 2001 WO
WO-01/30768 May 2001 WO
WO-01/53266 July 2001 WO
WO-01/57034 August 2001 WO
WO-01/81346 November 2001 WO
WO-01/81346 November 2001 WO
WO-03/035075 May 2003 WO
WO-03/106622 December 2003 WO
WO-03/106622 December 2003 WO
WO-2004/007491 January 2004 WO
WO-2004/012768 February 2004 WO
WO-2004/026285 April 2004 WO
WO-2004/026285 April 2004 WO
WO-2004/029055 April 2004 WO
WO-2004/052373 June 2004 WO
WO-2004/056820 July 2004 WO
WO-2004/089925 October 2004 WO
WO-2004/108708 December 2004 WO
WO-2004/108709 December 2004 WO
WO-2004/108713 December 2004 WO
WO-2004/108713 December 2004 WO
WO-2004/108715 December 2004 WO
WO-2004/108715 December 2004 WO
WO-2005/016348 February 2005 WO
WO-2005/016349 February 2005 WO
WO-2005/067901 July 2005 WO
WO-2005/067901 July 2005 WO
WO-05/113556 December 2005 WO
WO-05/120511 December 2005 WO
WO-2006/089106 August 2006 WO
WO-2009/058361 May 2009 WO
WO-2009/088986 July 2009 WO
WO-2010/065923 June 2010 WO
WO-2010/065923 June 2010 WO
WO-2010/123931 October 2010 WO
WO-2011/146882 November 2011 WO
WO-2014/023083 February 2014 WO
WO-2014/128612 August 2014 WO
WO-2015/010641 January 2015 WO
WO-2015/051241 April 2015 WO
WO-2015/081127 June 2015 WO
WO-2015/168079 November 2015 WO
WO-2015/191726 December 2015 WO
WO-2015/191743 December 2015 WO
WO-2015/191745 December 2015 WO
WO-2015/191752 December 2015 WO
WO-2015/191754 December 2015 WO
Other references
  • “Acute Congestive Heart Failure”, Thomas N. Levin, Postgraduate Medicine, vol. 101, No. 1, 1997.
  • Sutton, A. (Jun. 9, 2006). “Baylor, St. Luke's study uses gene therapy as pancreatic cancer”, located at <http: //www.bcm.edu/news/item.cfm?newsID=640>, last visited on Sep. 2, 2006, 2 pages.
  • Anonymous (2006). “Cardiovascular Disease: Treatment for Stroke”, Stanford Hospital & Clinics, located at <http://www.stanfordhospital.com/healthLib/atoz/cardiac/stktreat.html>, last visited on Sep. 19, 2006, 2 pages.
  • Marchione et al. (2006). “Drugs hold promise in kidney cancer fight”, located at <http://www.ledger-enquirer.com/mld/ledgerenquirer/living/health/14744763.htm> last visited on. Sep. 2, 2006, 3 pages.
  • Anonymous (2006). “Heart Disease”, WebMD, located at <http://www.webmd.com/content/pages/9/1675_57842.htm> as retrieved on Sep. 14, 2006, 1 page.
  • Anonymous, (2010). “Multiple Sclerosis”, located at <http://www.health.nytimes.com/health/guides/disease/multiple-sclerosis/overview.html> last visited Aug. 1, 2010, 4 pages.
  • Anonymous, (2004). “NIH Heart Disease & Stroke Research: Fact Sheet”, American Heart Association, located at <http://www.americanheart.org/presenter.jhtml?identifier=3010188>, last visited Feb. 17, 2004, 1 page.
  • Anonymous (2010). “Spinal Cord Injury”, located at <http://www.medicinenet.com/spinal_cord_injury/page.htm> last visited on Aug. 1, 2010, 3 pages.
  • Anonymous (2010) “Systemic Lupus Erythematosus”, located at <http://www.nlm.nih.gov/medlineplus/ency/article/000435.htm> last visited Aug. 1, 2010, 4 pages.
  • “Chemia Lekow,” ed. E. Pawelczyk, PZWL, Warszawa 1986, see, part 1.2.2.
  • “Preparatyka Organiczna”, ed. A.I. Vogel, WNT, Warszawa 1984, page, e.g. 83.
  • Abu-Duhier et al., Br. J. Haematol. (2001) 113:983-988.
  • Adamkiewicz, “Tumor Angiogenesis: Mechanisms” IMT Marburg—Research Group, retrieved from the internet on Apr. 13, 2004, located at: <http://www.imt.uni-marburg.de/˜adamkiew/mechanism.html>, 2 pages.
  • Advisory Action from U.S. Appl. No. 11/596,092, dated Jul. 27, 2010.
  • Ager et al., J. Med. Chem. (1977) 20:379-386.
  • Ali et al., Nature (2004) 431:1007-1011.
  • Alon et al., “The molecular basis of leukocyte adhesion to and migration through vascular endothelium,” Mirelman et al. (eds.), Life Sciences Open Day Book 2002, Weizmann Institute of Science, Life Sciences Department, Chapter 8, vol. 2:206-207 (2002), retrieved from the internet on Sep. 2, 2005, located at <http://www.weizmann.ac.il/Biology/open_day_2002/book/ronen_alon.pdf>, 2 pages.
  • Amendment from U.S. Appl. No. 09/841,341, filed Aug. 21, 2002.
  • Amendment from U.S. Appl. No. 10/027,591, filed Jun. 3, 2003.
  • Amendment in Response to Final Office Action from U.S. Appl. No. 11/596,092, filed Jul. 19, 2010.
  • Amendment in Response to Non-Final Office Action / Restriction Requirement from U.S. Appl. No. 11/884,566, filed Jun. 7, 2010.
  • Amendment in Response to Non-Final Office Action from U.S. Appl. No. 10/918,803, filed Oct. 1, 2008.
  • Amendment in Response to Non-Final Office Action from U.S. Appl. No. 10/918,803, filed Sep. 4, 2009.
  • Amendment in Response to Non-Final Office Action from U.S. Appl. No. 11/110,204, filed Dec. 31, 2008.
  • Amendment in Response to Non-Final Office Action from U.S. Appl. No. 11/110,204, filed Sep. 4, 2009.
  • Amendment in Response to Non-Final Office Action from U.S. Appl. No. 11/110,204, filed Jun. 4, 2010.
  • Amendment in Response to Non-Final Office Action from U.S. Appl. No. 11/596,092, filed Nov. 10, 2009.
  • Amendment in Response to Non-Final Office Action from U.S. Appl. No. 11/596,092, filed Mar. 24, 2010.
  • Amendment Under 37 C.F.R. § 1.111 from U.S. Appl. No. 11/129,006, filed Apr. 12, 2010.
  • Amendment Under 37 C.F.R. § 1.111/Restriction Requirement from U.S. Appl. No. 11/110,204, filed Apr. 10, 2008.
  • Amendment With Request for Continued Examination from U.S. Appl. No. 11/596,092, filed Sep. 1, 2010.
  • Amin et al., Circ Res (2003) 93(4):321-329.
  • Amine, M.S. et al. (Nov. 1998). “Uses of Quinazolin-2-[(β-Propionoyl) Isothiocyanate]-4-One as a Building Block in Synthesis of Some Heterocyclic Compounds of Expected Biological Activity,” Indian Journal of Chemistry 37B(11):1153-1156.
  • Angel, Activities of Phosphoinositide Kinase-3 (P13K) (1999) retrieved from the internet on May 22, 2003, URL: http://www.chem.csustan.edu/chem4400/SJBR/angel99.htm.
  • Angio World, “How Angiogenesis Complicates Psoriasis” (2001) retrieved from the internet on Apr. 13, 2004, located at <http://www.angioworld.com/psoriasis.htm>,1 page.
  • Annabi et al., J. Cell. Biochem. (2004) 91:1146-1158.
  • Aoki et al., PNAS USA (2001) 98:136-141.
  • Aoudjit et al., J. Immunol. (1998) 161:2333-2338.
  • Arcaro et al., Biochem. J. (1994) 298:517-520.
  • Asti et al., Pulm. Pharmacol. Ther. (2000) 13:61-69.
  • Ausprunk et al., Microvasc. Res. (1977) 14:53-65.
  • Australian Re-Examination Report dated Sep. 3, 2015, for Australian Patent No. 2001255667, filed Apr. 24, 2001, 7 pages.
  • Azenabor, A.A. et al. (2006). “Macrophage Antioxidant Enyzmes Regulate Chlamydia pneumoniaechronicity: Evidence of the Effect of Redox Balance on Host-Pathogen Relationship,” Immunobiology 211(5):325-339.
  • Bader, A.G. et al. (2005). “Oncogenic P13K Deregulates Transcription and Translation,” Nature Reviews Cancer 5(12):921-922 (abstract and introduction).
  • Barakat et al., Chemical Abstracts (1996) 124(21):1334.
  • Barakat, S.E-S. et al. (Dec. 1994). “Synthesis and CNS Depressant Activity of Some New Quinazoline Derivatives,” Az. J. Pharm. Sci. 14:239-246.
  • Bardet et al., 9th Congress of the European Hematology Association Geneva Palexpo, Switzerland, Jun. 10-13, 2004, View Abstract data, Abstract nr.: 620.
  • Barker, Lancet (1991) 338:227-230.
  • Benekli et al., Blood (2002) 99:252-257.
  • Benekli et al., Blood (2003) 101:2940-2954.
  • Bennett et al., Ann. Intern. Med. (1985) 103:620-625.
  • Bennett et al., J. Pharmacol. Exp. Ther. (1997) 280:988-1000.
  • Bergers et al., Science (1999) 284:808-812.
  • Bharadwaj et al., J. Immunol. (2001) 166:6735-6741.
  • Binetruy-Tournaire et al., EMBO J. (2000) 19:1525-1533.
  • Bloemen et al., Am. J. Respir. Crit. Care Med. (1996) 153:521-529.
  • Boehm et al., Nature (1997) 390:404-407.
  • Borregaard et al., Blood (1997) 89:3503-3521.
  • Boudewijn et al., Nature (1995) 376:599-602.
  • Bouscary et al., Blood (2003) 101:3436-3443.
  • Bouscary et al., Oncogene (2001) 20:2197-2204.
  • Bowes et al., Exp. Neurol. (1993) 119:215-219.
  • Brennan et al., Arthritis Res. (2002) 4(Suppl. 3):S177-S182.
  • Brown et al., 44th Annual Meeting of the American Society of Hematology, Philadelphia, PA, Dec. 6-10, 2002, Abstract No. 3012, p. 761A.
  • Brown, J. et al. (2010). “Clinical Activity in a Phase 1 Study of Cal-101, an Isoform-Selective Inhibitor of Phosphatidylinositol 3-Kinase P110Delta, in Patients with B-Cell Malignancies,” Haematologica 95(52):466, Abstract No. 1130.
  • Brunn et al., EMBO J. (1996) 15:5256-5267.
  • Burgering et al., Nature (1995) 376:599-602.
  • Butcher et al., Science (1996) 272:60-66.
  • Cadwallader et al., J. Immunol. (2002) 169:3336-3344.
  • Cantley et al., PNAS USA (1999) 96:4240-4245.
  • Cantley et al., Science (2002) 296:1655-1657.
  • Cardone et al., Science (1998) 282:1318-1321.
  • Carnero et al., FEB Letters (1998) 422:155-159.
  • CAS Abstract, Accession No. DN 86:83505 [1977] pp. 112-118.
  • Cebon et al., Cancer Immun. (2003) 3:7-25. I.
  • Chang et al., Exp. Opin. Ther. Patents (2001) 11:45-59.
  • Chang, BioMed. Eng. Online (2003) 2:12.
  • Chantry, D. et al. (1997). “p110δ, a Novel Phosphatidylinositol 3-Kinase Catalytic Subunit That Associates with p85 and Is Expressed Predominantly in Leukocytes,” J. Biol. Chem. 272(31):19236-19241.
  • Chapman-Kirkland, E.S. et al. (1991). “Superoxide Anion Production From Human Neutrophils Measured with an Improved Kinetic and Endpoint Microassay,” J Immunol Meth 142(1):95-104.
  • Chen et al., Blood (2000) 96:3181-3187.
  • Chern et al., Chem. Pharm. Bull. (1998) 46(6):928-933.
  • Chern et al., Chemical Abstracts (1998) 129(16):676.
  • Chopp et al., Stroke (1994) 25:869-876.
  • Choy et al., Arthritis & Rheumatism (2002) 46:3143-3150.
  • Clark et al., J. Neurosurg. (1991) 75:623-627.
  • Clavel et al., Joint Bone Spine (2003) 70:321-326.
  • Clayton et al., J. Exp. Med. (2002) 196:753-763.
  • Cleary, J.M. et al. (2010). “Development of Phosphoinositide-3 Kinase Pathway Inhibitors for Advanced Cancer,” Curr. Oncol. Rep. 12:87-94.
  • Coligan et al., Current Protocols in Protein Science (2002) 3:15-20.
  • Computer Search Cart Navigator, retrieved from the internet on Mar. 22, 2001, URL: http://www.chemnavigator.com/members/CartNavigator.asp#sample1, 8 pages.
  • Constantin et al., Immunity (2000) 13:759-769.
  • Cosimi et al., J. Immunol(1990) 144:4604-4612.
  • Coxon, Immunity (1996) 5:653-666.
  • Creamer et al., Angiogenesis (2002) 5:231-236.
  • Cross et al., Inflamm. Res. (1999) 48:255-261.
  • Curnock et al., Immunology (2002) 105:125-136.
  • Dahia et al., Hum. Mol. Genet. (1999) 8:185-193.
  • Dallegri et al., Inflamm. Res. (1997) 46:382-391.
  • Das et al., Prog. Retin. Eye Res. (2003) 22:721-748.
  • Datta et al., Cell (1997) 91:231-241.
  • Datta et al., Genes & Dev. (1999) 13:2905-2927.
  • Davies et al., Biochem. J. (2000) 351:95-105.
  • De Benedetti et al., Clin. Exper. Reheum. (1992) 10:493-498.
  • Deininger et al., Blood (2000) 96:3343-3356.
  • Demeester et al., Transplantation (1996) 62:1477-1485.
  • Descamps et al., J. Immunol. (2004) 173:4953-4959.
  • Doggett et al., Biophys. J. (2002) 83:194-205.
  • Domanig, R. (1981). “Chinazolinone, 2. Mitt: Synthese Und Einige Reaktionen Von 2-Azidomethyl-3-Aryl-4-Chinazolinonen,” Monatshefte für Chemie 112(10):1195-1202. (English translation of abstract only).
  • Dorland's Illustrated Medical Dictionary (2003), retrieved Oct. 21, 2005 from Xreferplus, http://www.xreferplus.com/entry/4196914.
  • Downward, Nature (1995) 376:553-554.
  • Drakesmith et al., Immunol. Today (2000) 21:214-217.
  • Druker et al., New England Journal of Medicine (2001) 344:1038-1042.
  • Dunne et al., Blood (2002) 99:336-341.
  • Edwards et al., Canc. Res. (2002) 62:4671-4677.
  • Eichholtz et al., J. Biol. Chem. (1993) 268:1982-1986.
  • El-Fattah et al., Indian J Hetercyclic Chemistry (1995) 4:199-202.
  • El-Feky, S.A. et al. (1985). “Synthesis of Certain New Sulfur-Containing Quinazolinone Derivatives Likely to Possess CNS Depressant Action,” Egyptian Journal of Pharmaceutical Sciences 24(1-4):39-47.
  • El-Feky et al., Chemical Abstracts (1987) 106(13):650.
  • El-Feky et al., Chemical Abstracts (1999) 131(23):497.
  • El-Feky, S.A. (Aug. 1998). “Novel Quinazolinones From 2-Cyanomethyl-3-Phenyl-4(3H) Quinazolinone,” Bollettino Chimico Farmaceutico 137(7):286-289.
  • Engelman et al., Nature Reviews (2006) 7:606-619.
  • Environmental Protection Agency, EPA-Radiation Information (EPA's Radiation Protection Program:Information) “Ionizing Radiation Fact Sheet Series No. 1” (May 1998) Retrieved on Apr. 21, 2004: http://www.epa.gov/radiation/docs/ionize/ionize.htm, 4 pages.
  • Erbagci et al., Clin. Biochem. (2001) 34:645-650.
  • Estey, Cancer (2001) 92:1059-1073.
  • Etzion I, Pediatr. Res. (1996) 39:191-198.
  • European Search Report dated Mar. 29, 2011, for EP Patent Application No. 10163434.3, filed on Apr. 24, 2001, 9 pages.
  • European Search Report dated Jun. 6, 2013 for EP Patent Application No. 13150110.8, filed May 12, 2005, 6 pages.
  • Extended European Search Report dated Dec. 10, 2013, for EP Patent Application No. 13150110.8, filed May 12, 2005, 10 pages.
  • Faffe et al., Eur. Respir. J. (2000) 15:85-91.
  • Fantl et al., Ann. Rev. Biochem. (1993) 62:453-481.
  • Faust et al., Blood (2000) 96:719-726.
  • Final Office Action from U.S. Appl. No. 10/918,803, dated Jan. 8, 2009.
  • Final Office Action from U.S. Appl. No. 11/129,006, dated Oct. 5, 2010.
  • Final Office Action from U.S. Appl. No. 11/596,092, dated May 18, 2010.
  • Final Office Action dated Oct. 24, 2011, for U.S. Appl. No. 12/732,128, filed Mar. 25, 2010, 8 pages.
  • Final Office Action dated Feb. 15, 2012, for U.S. Appl. No. 12/732,124, filed Mar. 25, 2010, 12 pages.
  • Final Office Action dated Jun. 7, 2012, for U.S. Appl. No. 11/129,006, filed May 12, 2005, 14 pages.
  • Final Office Action dated Jul. 9, 2013, for U.S. Appl. No. 13/399,828, filed Feb. 17, 2012, 6 pages.
  • First Preliminary Amendment from U.S. Appl. No. 12/538,748, filed Apr. 1, 2010.
  • Flinn, I.W. et al. (2009). “Preliminary Evidence of Clinical Activity in a Phase 1 Study of CAL-101, a Selective Inhibitor of the p1108 Isoform of Phosphatidylinositol 3-Kinase (P13K), in Patients with Select Hematologic Malignancies,” Journal of Clinical Oncology 27:156s, Abstract 3543.
  • Flinn, I.W. et al. (Nov. 20, 2009). “Evidence of Clinical Activity in a Phase 1 Study of CAL-101, an Oral P110Δ Isoform-Selective Inhibitor of Phosphatidylinositol 3-Kinase, in Patients with Relapsed or Refractory B-Cell Malignancies,” Blood 114(22):380, Abstract 922.
  • Flinn, W. et al. (Jun. 4-7, 2009). “Preliminary Evidence of Clinical Activity in a Phase 1 Study of CAL-101, A Potent Selective Inhibitor of the P110Delta Isoform of Phosphatidylinositol 3-Kinase, in Patients with B-Cell Maglignancies,” Haematologica 94(s2):303, Abstract 0744.
  • Folkman, Curr. Mol. Med. (2003) 3:643-651.
  • Folkman, Nat. Med. (1995) 1:27-31.
  • Fraser et al., Science (1991) 251:313-316.
  • Frey et al., Lancet (2008) 372(9643):1088-1099 (abstract).
  • Freyssinier et al., Br. J. Haematol. (1999) 106:912-922.
  • Fruman et al., Ann. Rev. Biochem. (1998) 67:481-507.
  • Fruman et al., Semin. Immunol. (2002) 14:7-18.
  • Furman, R.R. (Jul. 2010). “New Agents in Early Clinical Trials for CLL Therapy,” Clinical Advances in Hematology & Oncology 8(7):475-476.
  • Garcia-Barros et al., Science (2003) 300:1155-1159.
  • Genbank Accession No. AK040867, last updated Sep. 19, 2008, located at <http://www.ncbi.n1m.nih.gov.nuccore/26334014>, last visited on Apr. 16, 2010, 6 pages.
  • Genbank Accession No. AR255866, last updated Dec. 20, 2002, located at <http://www.ncbi.n1m.nih.gov.nuccore/27305059>, last visited on Apr. 16, 2010, 2 pages.
  • Genbank Accession No. BC035203, last updated Aug. 11, 2006, located at <http://www.ncbi.n1m.nih.gov/nuccore/23270986>, last visited on Apr. 16, 2010, 5 pages.
  • Genbank Accession No. NM_005026, last updated Apr. 11, 2010, located at <http://www.ncbi.n1m.nih.gov/nuccore/15654404>, last visited Apr. 16, 2010, 7 pages.
  • Genbank Accession No. NM_008840, last updated on Mar. 5, 2010, located at <http://www.ncbi.n1m.nih.gov/nuccore/255708435>, last visited on Apr. 16, 2010, 5 pages.
  • Genbank Accession No. U57843, last updated on May 9, 1997, located at <http://www.ncbi.nlm.nih.gov/nuccore/U57843>, last visited on Aug. 9, 2011, 2 pages.
  • Genbank Accession No. U86453, last updated on Jul. 7, 1998, located at <http://www.ncbi.n1m.nih.gov/nuccore/2317893>, last visited on Apr. 16, 2010, 3 pages.
  • Genbank Accession No. U86587, last updated Jul. 7, 1998, located at <http://www.ncbi.n1m.nih.gov/nuccore/2331237>, last visited on Apr. 16, 2010, 3 pages.
  • Genbank Accession No. XM_345606, last updated Jun. 22, 2006, located at <http://www.ncbi.n1m.nih.gov/nuccore/109475856?report=genbank>, last visited on Apr. 16, 2010, 3 pages.
  • Genbank Accession No. Y10055, last updated Oct. 7, 2008, located at <http://www.ncbi.n1m.nih.gov/nuccore/37496958>, last visited on Apr. 16, 2010, 3 pages.
  • Geng et al., Cancer Research (2001) 61:2413-19.
  • Geng et al., Cancer Research (2004) 64:4893-4899.
  • Geng et al., Cancer Research (2004) 64:8130-8133.
  • Gibson, (ed.), Antisense and Ribozyme Methodology, “Laboratory Companion” (1997) Table of Contents.
  • Gilliland et al., Blood (2002) 100:1532-1542.
  • Gilliland et al., Cancer Cell (2002) 1:417-420.
  • Gingras et al., Genes Dev. (2001) 15:2852-2864.
  • Gingras et al., Genes Dev. (2001) 15:807-826.
  • Glenjen et al., Int. J. Cancer (2002) 101:86-94.
  • Gorczynski et al., J. Immunol. (1994) 152:2011-2019.
  • Gorski et al., Cancer Research (1999) 59:3374-3378.
  • Gouilleux-Gruart et al., Blood (1996) 87:1692-1697.
  • Grant et al., Drugs of Today (2002) 38:783-791.
  • Green, S.J. et al. (1994). “Oxidative Metabolism of Murine Macrophages,” Chapter 14, Unit 14.5 in Current Protocols in Immunology, vol. 3, John Wiley & Sons, Inc., pp. 14.5.1-14.5.11.
  • Gross et al., Science (1998) 281:703-706.
  • Gu et al., Mol. Cell. Biol. (2000) 20:7109-7120.
  • Gupta et al., Int'l J Radiation Oncology Biology Physics (2003) 56(3):846-853.
  • Gute et al., Mol. Cell. Biochem. (1998) 179:169-187.
  • Guzman et al., Blood (2001) 98:2301-2307.
  • Guzman et al., Proc. Natl. Acad. Sci. (USA) (2002) 99:16220-16225.
  • Hadden, Int. Immunopharmacol. (2003) 3:1061-1071.
  • Hallahan et al., Proc. Natl. Acad. Sci (USA) (1997) 94:6432-6437.
  • Halloran et al., Arthritis Rheum. (1996) 39:810-819.
  • Hanamoto et al., Am. J. Pathol. (2004) 164(3):997-1006.
  • Hannigan et al., Proc. Natl. Acad. Sci. U.S.A. (2002) 99:3603-3608.
  • Hardma et al. (eds.), Goodman and Gilman's The Pharmacological Basis of Therapeutics (1996) 9th ed., pp. 11-16.
  • Harlan, Haematology 96, the Education Program Book of the 26th Congress of the International Society of Haematology. Singapore, 1996.
  • Harning et al., Transplantation (1991) 52:842-845.
  • Hartley et al., Cell (1995) 82:849-856.
  • Hartman et al., Cardiovasc. Res. (1995) 30:47-54.
  • Hasagawa et al., Int. Immunol. (1994) 6:831-838.
  • Hassan et al., Chinese Journal of Chemistry (1991) 9:262-269.
  • Hattori, H. et al. (May/Jun. 2010). “Reactive Oxygen Species as Signaling Molecules in Neutrophil Chemotaxis,” Communicative and Integrative Biology 3(3):278-281. _.
  • He et al., Opthalmol. Vis. Sci. (1994) 35:3218-3225.
  • Healy et al., Hum. Reprod. Update (1998) 4:736-740.
  • Healy et al., Pharma. Res. (Dec. 2004) 21:2234-2246.
  • Heit et al., J. Cell Biol. (2002) 159:91-102.
  • Hellman, Cancer: Principles and Practice of Oncology (1993) 4th ed., vol. 1:248-275.
  • Herman, S.E.M. et al. (Sep. 23, 2010). “Phosphatidylinositol 3-Kinase-δ Inhibitor CAL-101 Shows Promising Preclinical Activity in Chronic Lymphocytic Leukemia by Antagonizing Intrinsic and Extrinsic Cellular Survival Signals,” Blood 116(12):2078-2088.
  • Herold et al., Cell Immunol. (1994) 157:489-500.
  • Higuchi, Prodrugs as Novel Delivery Systems, vol. 14, ASCD Symposium Series, and in Roche (ed.), Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press (1987) Chapter 1, pp. 1-12.
  • Hilbert et al., J. Exper. Med. (1995) 182:243-248.
  • Hiles et al., Cell (1992) 70:419-429.
  • Hilmas et al., Rad. Res. (1975) 61:128-143.
  • Hirsch et al., Science (2000) 287:1049-1053.
  • Horgan et al., Am. J. Physiol. (1991) 261:H1578-H1584.
  • Hsieh, S.N. (2003). “Identification of Pl3K≢ in Endothelial Cells and Its Involvement in Sphingosine 1-Phosphate Mediated Endothelial Cell Migration,” Dissertation, Friedrick Schiller University, Jena, Germany, 104 pages.
  • Hu et al., Mol. Cell. Biol. (1993) 13:7677-7688.
  • Hu et al., Science (1995) 268:100-102.
  • Hunter, Cell (1995) 83:1-4.
  • Hussong et al., Blood (2000) 95:309-313.
  • Ikeda, H. et al. (Feb. 2009). “CAL-101: A Selective Inhibitor of P13K p110δ for the Treatment of Multiple Myeloma,” Clinical Lymphoma and Myeloma 9(Supp. 1):S98-S99.
  • Ikeda, H. et al. (Nov. 16, 2008). “CAL-101, a Specific Inhibitor of the p110δ Isoform of Phosphatidylinositide 3-Kinase Induces Cytotoxicity in Multiple Myeloma (MM),” Blood 112(11):950, Abstract No. 2753.
  • Ikeda, H. et al. (Sep. 2, 2010). “Pl3K/p110δ is a Novel Therapeutic Target in Multiple Myeloma,” Blood 116(9):1460-1468.
  • International Preliminary Report on Patentability for PCT/US2006/005621, dated Aug. 21, 2007, 8 pages.
  • International Search Report for International (PCT) Patent Application Serial No. PCT/US2004/026436, dated Dec. 2, 2004.
  • International Search Report for International (PCT) Patent Application Serial No. PCT/US2004/029561, dated May 25, 2005.
  • International Search Report for International (PCT) Patent Application Serial No. PCT/US2004/026834, dated Nov. 29, 2004.
  • International Search Report for International (PCT) Patent Application Serial No. PCT/US2004/037860, dated May 6, 2005.
  • International Search Report dated Aug. 29, 2005, for PCT Application No. PCT/US2005/016778, filed on May 12, 2005, 4 pages.
  • International Search Report dated Sep. 15, 2006 for PCT Application No. PCT/US2006/005621, filed on Feb. 16, 2006, 4 pages.
  • International Search Report dated Sep. 29, 2015 for PCT Application No. PCT/US2015/035126, Internationally filed on Jun. 10, 2015, 4 pages.
  • Interview Summary from U.S. Appl. No. 10/918,825, dated Jun. 14, 2006.
  • Ishida-Okawara, A. et al. (Dec. 12, 1996). “Modulation of Degranulation and Superoxide Generation in Human Neutrophils by Unsaturated Fatty Acids of Odd Carbon Numbers,” BioChimica et Biophysica Acta 1314(3):239-246.
  • Ismail and Sayed, Indian Journal of Chemistry (1982) 21B(5):461-462.
  • Ismail et al., Chemical Abstracts (1983) vol. 98, No. 1, p. 406.
  • Isobe et al., Science (1992) 255:1125-1127.
  • Johnson et al., Intl. J. Rad. One. Biol. Phys. (1976) 1:659-670.
  • Johnson et al., J. Endourol. (2003) 17:557-562.
  • Jordan, Nature Reviews: Drug Discovery (2003) 2:205.
  • Jou et al., Mol. Cell. Biol. (2002) 22:8580-8591.
  • Kahl, B.S. (May 2010). “Novel Agents for Non-Hodgkin Lymphoma,” Clinical Advances in Hematology & Oncology 8 (5)(Suppl. 10):10-15.
  • Kakimoto et al., Cell. Immunol. (1992) 142:326-337.
  • Kallman et al., Canc. Res. (1972) 32:483-490.
  • Kandel et al., Exp. Cell Res. (1999) 253:210-229.
  • Kawasaki et al., J. Immunol. (1993) 150:1074-1083.
  • Kim et al., Endocrin. (2000) 141:1846-1853.
  • Kim, Retrieved from the Internet on Apr. 13, 2004: URL: http://www.math.umn.edu/˜yjkim/biopaper/timy,html.
  • Kishimoto et al., Cell (1987) 50:193-202. I.
  • Klein et al., Cell. Signal. (2001) 13:335-343.
  • Klippel et al., Mol. Cell. Biol. (1994) 14:2675-2685. I.
  • Knall et al., Proc. Natl. Acad. Sci. (USA) (1997) 94:3052-3057.
  • Knight and Shokat, Chemistry and Biology (2005) 12:621-637.
  • Knight et al., Bioorganic & Medicinal Chemistry (Jul. 2004) 12:4749-4759.
  • Knoerzer et al., Toxicol. Pathol. (1997) 25:13-19.
  • Kolonin et al., Nature Medicine (2004) 10:625-632.
  • Kong et al., J. Biol. Chem. (2000) 275:36035-36042.
  • Kopf et al., Nature (1994) 368:339-342.
  • Krugmann et al., J. Biol. Chem. (1999) 274:17152-17158.
  • Kumar et al., Blood (2003) 101(10):3960-3968.
  • Kunkel et al., Circ. Res. (1996) 79:1196-1204.
  • Lannutti, B.J. et al. (Apr. 2009). “CAL-101, a Specific Pl3K p110δ Inhibitor for the Treatment of Hematological Maglignancies,” Proceedings of the American Association for Cancer Research 50:1400, Abstract No. #SY32-2.
  • Lannutti, B.J. et al. (Nov. 16, 2008). “CAL-101, a Potent Selective Inhibitor of the p110d Isoform of Phosphatidylinositol 3-Kinase, Attenuates PI3K Signaling and Inhibitos Proliferation and Survival of Acure Lumpoblastic Leukemia in Addition to a Range of Other Hematological Malignancies,” Blood 112(11):12, Abstract No. 16.
  • Lannutti, B.J. et al. (Nov. 20, 2009). “CAL-101, An Oral P110δ Selective Phosphatidylinositol-3-Kinase (PI3K) Inhibitor for the Treatment of B Cell Malignancies Inhibits PI3K Signaling, Cellular Viability and Protective Signals of the Microenvironment,” Blood 114(22):120-121, Abstract No. 286.
  • Lannutti, J. et al. (2010). “Demonstration of Pharmacodynamic Target Inhibition and Chemokine Modulation in Patients with CLL Following Treatment with CAL-101, a Selective Inhibitor of the P110 Delta Isoform of PI3K,” Haematologica 95(52):45-46, Abstract No. 0113.
  • Lannutti, J. et al. (Jun. 4-7, 2009). “CAL-101, A Specific Inhibitor of the P11-Delta Isoform of Phosphatidylinositide 3-Kinase, for the Treatment of Non-Hodgkins Lymphomas,” Haematologica 94(S2):272-273, Abstract No. 0668.
  • Lecoq-Lafon et al., Blood (1999) 93:2578-2585.
  • Lemmon et al., Trends Cell. Biol. (1997) 7:237-242.
  • Letter From Polish Patent Law Firm “Patpol” translating Office Action from Polish Patent Application No. P-358590, dated Feb. 27, 2008.
  • Li et al., Trends Biochem. Sci. (Jan. 2004) 29:32-38.
  • Liang et al., Molecular Cancer Therapeutics (2003) 2(4):353-360.
  • Liekens et al., Biochem. Pharmacol. (2001) 61:253-270.
  • Liu et al., J. Immunol. (Jan. 2004) 172:7-13.
  • Lowell et al., J. Cell Biol. (1996) 133:895-910.
  • Luo et al., Cancer Cell (2003) 4:257-262.
  • Luo, et al., Leukemia (2003) 17:1-8.
  • Luster, N. Engl. J. Med. (1998) 338:436-445.
  • Madge et al., J. Biol. Chem. (2000) 275:15458-15465.
  • Manning et al., Mol. Cell (2002) 10:151-162.
  • Marley et al., Br. J. Haematol. (May 2004) 125:500-511.
  • May, S.E. et al. (Nov. 16, 2008). “CAL-101, a Selective Inhibitor of the p110δ Isoform of Phosphatidylinositol 3-Kinase, Effectively Induces Apoptosis in Primary Chronic Lumphocytic Leukemia Cells Providing a Novel Therapeutic Strategy for the Treatment of this Disease,” Blood 112(11):1085-1086, Abstract No. 3165.
  • Meneses et al., Gene Ther. (2001) 8:646-648. I.
  • Milella et al., J. Clin. Invest. (2001) 108:851-859.
  • Miller et al., Nucleic Acids Res. (1988) 16:1215.
  • Moehler et al., Ann. Hematol. (2001) 80:695-705.
  • Moore, J. Clin. Invest. (2002) 109:313-315.
  • Moulton et al., Circ. (1999) 99:1726-1732.
  • Mulligan et al., J. Immunol. (1995) 154:1350-1363.
  • Mulligan et al., Proc. Natl. Acad. Sci. (USA) (1993) 90:11523-11527.
  • Nagase et al., Am. J. Respir. Crit. Care Med. (1996) 154:504-510.
  • Nakao et al., Leukemia (1996) 10:1911-1918.
  • Nakao et al., Muscle Nerve (1995) 18:93-102.
  • Neshat et al., Proc. Natl. Acad. Sci. (USA) (2001) 98:10314-10319.
  • Ninomiya et al., J. Biol. Chem. (1994) 269:22732-22737.
  • Non-Final Office Action from U.S. Appl. No. 11/596,092, dated Dec. 24, 2009.
  • Non-Final Office Action from U.S. Appl. No. 09/841,341, dated Apr. 25, 2002.
  • Non-Final Office Action from U.S. Appl. No. 10/027,591, dated Feb. 26, 2003.
  • Non-Final Office Action from U.S. Appl. No. 10/918,803, dated Apr. 1, 2008.
  • Non-Final Office Action from U.S. Appl. No. 10/918,803, dated Mar. 16, 2010.
  • Non-Final Office Action from U.S. Appl. No. 10/918,825, dated Nov. 7, 2005.
  • Non-Final Office Action from U.S. Appl. No. 11/110,204, dated Aug. 5, 2008.
  • Non-Final Office Action from U.S. Appl. No. 11/110,204, dated Feb. 4, 2010.
  • Non-Final Office Action from U.S. Appl. No. 11/110,204, dated Jun. 17, 2009.
  • Non-Final Office Action from U.S. Appl. No. 11/129,006, dated Dec. 15, 2009.
  • Non-Final Office Action dated Jan. 20, 2012 for U.S. Appl. No. 13/163,597, filed Jun. 17, 2011, 14 pages.
  • Non-Final Office Action dated Oct. 17, 2011 for U.S. Appl. No. 12/732,124, filed Mar. 25, 2010, 8 pages.
  • Non-Final Office Action from U.S. Appl. No. 11/596,092, dated Jun. 10, 2009.
  • Non-Final Office Action from U.S. Appl. No. 11/884,566, dated Aug. 3, 2010.
  • Non-Final Office Action dated Jun. 28, 2011, for U.S. Appl. No. 12/732,124, filed Mar. 25, 2010, 11 pages.
  • Non-Final Office Action dated Aug. 2, 2012, for U.S. Appl. No. 12/575,277, filed Oct. 7, 2009, 8 pages.
  • Non-Final Office Action dated Aug. 7, 2012, for U.S. Appl. No. 12/575,367, filed Oct. 7, 2009, 9 pages.
  • Non-Final Office Action dated Feb. 13, 2013, for U.S. Appl. No. 13/247,962, filed Sep. 28, 2011, 21 pages.
  • Non-Final Office Action dated Mar. 1, 2013, for U.S. Appl. No. 13/399,828, filed Feb. 17, 2012, 6 pages.
  • Non-Final Office Action dated Mar. 25, 2013, for U.S. Appl. No. 13/728,807, filed Dec. 27, 2012, 13 pages.
  • Non-Final Office Action dated Jun. 26, 2013, for U.S. Appl. No. 13/765,610, filed Feb. 12, 2013, 9 pages.
  • Non-Final Office Action dated Jun. 25, 2014, for U.S. Appl. No. 14/049,154, filed Oct. 8, 2013, 15 pages.
  • Non-Final Office Action dated Jun. 25, 2014, for U.S. Appl. No. 14/049,163, filed Oct. 8, 2013, 15 pages.
  • Non-Final Office Action dated Feb. 3, 2015, for U.S. Appl. No. 14/284,331, filed May 21, 2014, 16 pages.
  • Non-Final Office Action dated Oct. 8, 2015, for U.S. Appl. No. 14/323,925, filed Jul. 3, 2014, 8 pages.
  • Non-Final Office Action dated Nov. 16, 2015, for U.S. Appl. No. 14/092,287, filed Nov. 27, 2013, 11 pages.
  • Notice of Allowance from U.S. Appl. No. 09/841,341, dated Oct. 7, 2002.
  • Notice of Allowance from U.S. Appl. No. 10/027,591, dated Jul. 29, 2003.
  • Notice of Allowance from U.S. Appl. No. 10/337,192, dated Mar. 11, 2004.
  • Notice of Allowance from U.S. Appl. No. 10/697,912, dated Dec. 30, 2004.
  • Notice of Allowance dated Nov. 8, 2010, for U.S. Appl. No. 11/110,204, filed Apr. 20, 2005, 6 pages.
  • Notice of Allowance dated Jun. 26, 2012 for U.S. Appl. No. 12/732,124, filed Mar. 25, 2010, 6 pages.
  • Notice of Allowance dated Nov. 13, 2012 for U.S. Appl. No. 12/732,124, filed Mar. 25, 2010, 11 pages.
  • Notice of Allowance dated Feb. 21, 2013 for U.S. Appl. No. 12/575,367, filed Oct. 7, 2009, 5 pages.
  • Notice of Allowance dated May 20, 2013, for U.S. Appl. No. 13/730,276, filed Dec. 28, 2012, 7 pages.
  • Notice of Allowance dated Jul. 8, 2013 for U.S. Appl. No. 13/728,807, filed Dec. 27, 2012, 9 pages.
  • Notice of Allowance dated Jul. 8, 2013, for U.S. Appl. No. 13/730,256, filed Dec. 28, 2012, 9 pages.
  • Notice of Allowance dated Aug. 28, 2013, for U.S. Appl. No. 12/575,277, filed Oct. 7, 2009, 6 pages.
  • Notice of Allowance dated Sep. 19, 2013, for U.S. Appl. No. 13/399,828, filed Feb. 17, 2012, 6 pages.
  • Notice of Allowance dated Oct. 3, 2013, for U.S. Appl. No. 13/247,962, filed Sep. 28, 2011, 9 pages.
  • Notice of Allowance dated Oct. 18, 2013, for U.S. Appl. No. 13/765,610, filed Feb. 12, 2013, 10 pages.
  • Notice of Allowance dated Feb. 21, 2014, for U.S. Appl. No. 13/765,610, filed Feb. 12, 2013, 9 pages.
  • Notice of Allowance dated Oct. 9, 2014, for U.S. Appl. No. 14/049,154, filed Oct. 8, 2013, 8 pages.
  • Notice of Allowance dated Oct. 10, 2014, for U.S. Appl. No. 14/049,163, filed Oct. 8, 2013, 8 pages.
  • Notice of Allowance dated May 14, 2015, for U.S. Appl. No. 14/284,331, filed May 21, 2014, 9 pages.
  • Notice of Reexamination for Chinese Patent Application No. 0811654.X, dated Nov. 5, 2009; 7 pages.
  • Notice Regarding Non-Compliant Amendment for U.S. Appl. No. 10/918,803, dated Nov. 19, 2009, 3 pages.
  • Notification of Reasons for Rejection for Japanese Patent Application No. 2003-537642, dated May 26, 2009, 4 pages.
  • Office Action for European Patent Application No. 04 816 855.3, dated Oct. 21, 2008, 4 pages.
  • Office Action for European Patent Application No. 05 752 122.1, dated Mar. 25, 2013, 4 pages.
  • Office Action for European Patent Application No. 05752122.1, dated Nov. 5, 2014, 3 pages.
  • Ohno-Matsui et al., Invest. Ophthalmol. Vis: Sci. (2003) 44:5370-5375.
  • Okkenhaug et al., Science (2002) 297:1031-1034.
  • Oppenheimer-Marks et al., J. Clin. Invest. (1998) 101:1261-1272.
  • Oshiro et al., Stroke (1997) 28:2031-2038.
  • Otsu et al., Cell (1991) 65:91-104.
  • Paez et al., Frank (ed.), Cancer Treatment and Research (2003) 115:146 Kluwer Academic Publishers.
  • Pages et al., Nature (1994) 369:327-329.
  • Palanki, Curr. Med. Chem. (2002) 9:219-227.
  • Paleolog et al., Angiogenesis (1998/1999) 2:295-307.
  • Panayotou et al., Trends in Cell Biol. (1992) 2:358-360.
  • Panes et al., Gastroenterology (1995) 108:1761-1769.
  • Parasharya and Parikh, J. Inst. Chemists (1992) 64(5):184-185.
  • Parasharya et al., Chemical Abstracts (1994) vol. 121, No. 9, p. 1065.
  • Park, S. et al. (2010). “Role of the PI3K/AKT and mTOR Signaling Pathways in Acute Myeloid Leukemia,” Haematologica 95(5):819-829.
  • Parker, Current Biology (1995) 5:577-579.
  • Passegue et al., Proc. Natl. Acad. Sci., (USA) (2003) 100 Supp. 1:11842-11849.
  • Patani, G.A. et al. (1996), “Bioisosterism: A Rational Approach in Drug Design,” Chem Rev. 96(8):3147-3176.
  • Pierce et al., J. Biol. Chem. (1997) 272:21096-21103.
  • Plows et al., J. Immunol. (1999) 162(2):1018-1023.
  • Podsypanina et al., Proc. Natl. Acad. Sci. (USA) (2001) 98:10320-10325.
  • Psychoyos et al., J. Immunol. Methods (1991) 137:37-46.
  • Puri et al., Blood (2005) 106(1):150-157, 144.
  • Puri et al., Blood (May 2004) 103:3448-3456.
  • Puri, K. et al. (Jul. 18-23, 2004). “A Role for Phosphoinositide 3-Kinase δ in Neutrophil Trafficking,” Immunology 2004: Cytokine Network, Regulatory Cells, Signaling, and Apoptosis: Collection of Free Papers Presented at the 12th International Congress of Immunology and 4th Annual Conference of FOCIS Medimond International Proceedings in Montreal, Canada on Jul. 18, 23, 2004, pp. 303-307.
  • Quirici et al., Br. J. Haematol. (2001) 115:186-194.
  • Rada, B.K. et al. (Nov. 1, 2004, e-published Jul. 13, 2004). “Dual Role of Phagocytic NADPH Oxidase in Bacterial Killing,” Blood 104(9):2947-2953.
  • Rameh et al., Cell (1995) 83:821-830.
  • Rameh et al., J. Biol. Chem. (1999) 274:8347-8350.
  • Rathman et al., J. Org. Chem. (1980) 45:2169-2176.
  • Remington'S Pharmaceutical Sciences (1990) 18th Ed., Chapter 89, pp. 1435-1712 Table of Contents Only.
  • Ren et al., Curr. Drug Targets Inflamm. Allergy (2003) 2(3):242-256.
  • Request for Continued Examination and Amendment Under 37 C.F.R. § 1.116 from U.S. Appl. No. 10/918,803, filed May 7, 2009.
  • Response to Election of Species Requirement from U.S. Appl. No. 10/918,803, filed Jun. 27, 2007.
  • Response to Non-Final Office Action from U.S. Appl. No. 10/918,803, filed Dec. 18, 2009, 7 pgs.
  • Response to Non-Final Office Action filed on Sep. 16, 2010, for U.S. Appl. No. 10/918,803, filed Aug. 13, 2004, 25 pages.
  • Response to Restriction Requirement from U.S. Appl. No. 10/918,803, filed Jan. 4, 2008, 4 pgs.
  • Response to Restriction Requirement from U.S. Appl. No. 11/129,006, filed May 12, 2009, 5 pgs.
  • Response to Restriction Requirement from U.S. Appl. No. 11/137,901, filed Feb. 6, 2008, 6pgs.
  • Response to Restriction Requirement from U.S. Appl. No. 11/596,092, filed May 27, 2009, 4pgs.
  • Response to Rule 312 Communication dated Oct. 4, 2012, for U.S. Appl. No. 12/732,124, filed Mar. 25, 2010, 7 pages.
  • Restriction Requirement from U.S. Appl. No. 10/918,803, dated Jun. 12, 2009.
  • Restriction Requirement from U.S. Appl. No. 10/918,803, dated Mar. 13, 2007.
  • Restriction Requirement from U.S. Appl. No. 10/918,803, dated Sep. 7, 2007.
  • Restriction Requirement from U.S. Appl. No. 11/110,204, dated Mar. 10, 2008.
  • Restriction Requirement from U.S. Appl. No. 11/129,006, dated Nov. 12, 2008.
  • Restriction Requirement from U.S. Appl. No. 11/137,901, dated Aug. 6, 2007.
  • Restriction Requirement from U.S. Appl. No. 11/137,901, dated May 23, 2008.
  • Restriction Requirement from U.S. Appl. No. 11/596,092, dated Jan. 28, 2009.
  • Restriction Requirement from U.S. Appl. No. 11/884,566, dated Apr. 5, 2010.
  • Restriction Requirement dated Oct. 14, 2010, for U.S. Appl. No. 12/732,124, filed Mar. 25, 2010, 9 pages.
  • Restriction Requirement dated Dec. 1, 2011, for U.S. Appl. No. 13/163,597, filed Jun. 17, 2011, 7 pages.
  • Restriction Requirement dated Jun. 7, 2012, for U.S. Appl. No. 13/399,828, filed Feb. 17, 2012, 5 pages.
  • Restriction Requirement dated Jul. 17, 2012, for U.S. Appl. No. 13/247,962, filed Sep. 28, 2011, 27 pages.
  • Restriction Requirement dated Sep. 11, 2012, for U.S. Appl. No. 13/399,828, filed Feb. 17, 2012, 7 pages.
  • Restriction Requirement dated May 8, 2015, for U.S. Appl. No. 14/092,287, filed Nov. 27, 2013, 8 pages.
  • Reyes et al., J. Clin. Invest. (2002) 109:337-346.
  • Rickert et al., Trends Cell Biol. (2000) 10:466-473.
  • Riesterer, Int'l J Radiation Oncology Biology Physics (2004) 361-368.
  • Roberts et al., Immunity (1999) 10:183-196.
  • Rodrigues et al., Mol. Cell. Biol. (2000) 20:1448-1459.
  • Rodriguez-Viciana et al., EMBO J. (1996) 15:2442-2451.
  • Roth et al., J. Immunol. Methods (1995) 188:97-116.
  • Rudd, Immunity (1996) 4:527-534.
  • Rupnick et al., Proc. Nat'l. Acad. Sci. (USA) (2002) 99:10730-35.
  • Sadhu et al., J. Immunol. (2003) 170:2647-2654.
  • Salven et al., Blood (1999) 94:3334-3339.
  • Salvesen et al., Cell (1997) 91:443-446.
  • Sasaki et al., Science (2000) 287:1040-1046.
  • Sauder et al., J. Am. Acad. Dermatol. (2002) 47:535-541.
  • Schimmer et al., J. Immunol. (1998) 160:1466-1471.
  • Schuch et al., Blood (2002) 100:4622-4628.
  • Schueneman et al., Canc. Res. (2003) 63:4009-4016.
  • Second Preliminary Amendment and Response to Notice to File Missing Parts of Nonprovisional Application from U.S. Appl. No. 12/575,277, filed Jan. 20, 2010.
  • Second Preliminary Amendment and Response to Notice to File Missing Parts of Nonprovisional Application from U.S. Appl. No. 12/575,367, filed Jan. 20, 2010.
  • Second Preliminary Amendment from U.S. Appl. No. 11/110,204, filed Aug. 24, 2007.
  • Second Preliminary Amendment from U.S. Appl. No. 11/884,566, filed May 13, 2008.
  • Sengupta et al., Circulation (2003) 107:2955-2961.
  • Shimamoto et al., Leukemia Res. (2003) 27:783-788.
  • Shiojima et al., Circ. Res. (2002) 90:1243-1250.
  • Shvidel et al., Hematol. J. (2002) 3:32-37.
  • Smith et al., Am. J. Respir. Cell Mol. Biol. (1996) 15(6):693-702.
  • Song et al., Canc. Res. (1974) 34:2344-2350.
  • Springer, Cell (1994) 76:301-314.
  • Stanovnik, B. et al. “The Tautomerism of Heterocycles: Substituent Tautomerism of Six-Membered Ring Heterocycles”, Advances in Heterocyclic Chemistry (2002) 91: 1-134. _.
  • Stein et al., Mol. Med. Today (2000) 6:347-357.
  • Stenmark et al., J. Cell. Sci. (1999) 112:4175-4183.
  • Stennicke et al., Biochim. Biophys. Acta. (2000) 1477:299-306.
  • Stephens et al., Current Biology (1994) 4:203-214.
  • Stirewalt et al., Nat. Rev. Cancer (2003) 3:650-665.
  • Stoyanov et al., Science (1995) 269:690-693.
  • Su et al.,Cancer Research (2003) 63:3585-3592.
  • Sumariwalla et al., Arthritis Res. Ther. (2002) 5:R32-R39.
  • Sunil et al., Respir. Res. (2002) 3:21.
  • Supplemental Amendment from U.S. Appl. No. 11/110,204, filed Oct. 27, 2009.
  • Supplemental Notice of Allowance from U.S. Appl. No. 10/337,192, dated Jun. 29, 2004.
  • Tager et al., J. Exp. Med. (2000) 192:439-446.
  • Talento et al., Transplantation (1993) 55:418-422.
  • Tamiya et al., Immunopharmacology (1995) 29:53-63.
  • Tan et al., Cancer Research (2003) 63:7663-7667.
  • Tan et al., J. Immunot Meths. (2000), 238:59-68.
  • Tan, J. et al. (Sep. 1, 2004). “A Specific Antagonist of the p110-Delta Catalytic Component of P13 Kinase, IC486068, Enhances Radiation-Induced Tumor Vascular Destruction,” International Journal of Radiation: Oncology Biology Physics 60(1):S195.
  • Tanaka et al., J. Immunol. (1993) 151:5088-5095.
  • Tang et al., J. Biol. Chem. (1999) 274:16741-16746.
  • Taylor et al., Curr. Opin. Rheumatol. (2005) 17(3):293-298.
  • Tesar et al., Med. Sc. Monit. (2002) 8:BR24-BR29.
  • The Merck Manual on “arthritis” (2008).
  • The Merck Manual on “rheumatoid arthritis” (2008).
  • The Merck Manual, 17th ed, (1999) p. 1001.
  • Thelan et al., Proc. Natl. Acad. Sci. (USA) (1994) 91:4960-4964.
  • Ting et al., Int. J. Rad. Biol. (1991) 60:335-339.
  • Vacca et al., Blood (1999) 9:3064-3073.
  • Van Dijk et al., Blood (2000) 96:3406-3413.
  • Van Eeden, S.F. et al. (Dec. 17, 1999). “The Use of Flow Cytometry to Measure Neutrophil Function,” Journal Immunol Meth 232:23-43.
  • Vanhaesebroeck et al., FASEB J. (1996) 10:A1395, Abst. No. 2280.
  • Vanhaesebroeck et al., Proc. Natl. Acad. Sci., (USA) (1997) 94:4330-4335.
  • Vanhaesebroeck et al., TIBS (1997) 22:267-272.
  • Vermes et al., J. Immunot Meth. (1995) 184:39-51.
  • Vippagunta, S.R. et al. (2001). “Crystalline Solids,” Advanced Drug Delivery 48:3-26.
  • Vivanco et al., Nat. Rev. Cancer (2002) 2:489-501.
  • Vlahos et al., J. Immunol. (1995) 154:2413-2422.
  • Volinia et al., EMBO J. (1995) 14:3339-3348.
  • Volinia et al., Genomics (1994) 24:472-477.
  • Volinia et al., Oncogene (1992) 7:789-793.
  • Webb, H.K. et al. (Apr. 2009). “CAL-101, a Potent and Selective Inhibitor of the Class 1 Phosphatidylinositol 3 Kinase (PI3K) p110δ: Preclinical Summary,” Proceedings of the American Association for Cancer Research 50:894-895, Abstract No. #3703.
  • Wegner et al., Lung (1992) 170:267-279.
  • Wegner et al., Science (1990) 247:456-459. I.
  • Weiner et al., Nat. Cell Biol. (1999) 1:75-81.
  • Weyand et al., Arthritis & Rheumatism (2000) 43:1041-1048.
  • Williams, D.A. et al. (2002). Foye's Principles of Medicinal Chemistry, Lippincott, Williams & Wilkins, Baltimore MD, Fifth Edition, pp. 50 and 59-61.
  • Williams, Methods Mol. Med. (2004) 98:207-216.
  • Williams et al., Chem. Biol. (2010) 17:123-134.
  • Wolff, ed., Burger's Medicinal Chemistry and Drug Discovery, 5th edition (1996) vol. 1, New York: John Wiley & Sons, pp. 975-977.
  • Written Opinion dated Sep. 29, 2015 for Application No. PCT/US2015/035126, Internationally filed on Jun. 10, 2015, 6 pages.
  • Wymann et al., Biochem. Biophys. Acta. (1998) 1436:127-150.
  • Wymann et al., Biochem. J. (1994) 298:517-520.
  • Wymann et al., Trends Immunol. Today (2000) 21:260-264.
  • Xing et al., Am. J. Pathol. (1993) 143:1009-1015.
  • Xu et al., Blood (2003) 102:972-980.
  • Yamasawa et al., Inflammation (1999) 23:263-274.
  • Yamaura et al., Int. J. Rad. Biol. (1976) 30:179-187.
  • Yao et al., Science (1995) 267:2003-2006. I.
  • Yum et al., J. Immunol. (2001) 167:6601-6608.
  • Zeng et al., Transplantation (1994) 58:681-689.
  • Zhao et al., Leukemia (2004) 18:267-75.
  • Zu, Y-L et al. (1998). “p38 Mitogen-Activated Protein Kinase Activation is Required for Human Neutrophil Function Triggered by TNF-α or FMLP Stimulation”, J Immunol 160:1982-1989.
  • Final Office Action dated Feb. 26, 2016, for U.S. Appl. No. 14/092,287, filed Nov. 27, 2013, 6 pages.
  • Chemical Encyclopedia (1988), publisher “Soviet encyclopedia”, Moscow, 2:365-369.
  • Engelman, et al. (Aug. 2009). “Targeting PI3K Signaling in Cancer: Opportunities, Challenges and Limitations”, Nat. Rev. Cancer 9:550-562.
  • Foster, A. (1984). “Deuterium Isotope Effects in Studies of Drug Metabolism”, Trends Pharmacol. Sci., 5(12):524-527.
  • International Preliminary Report dated Dec. 12, 2016, for PCT Patent Application No. PCT/US2015/035126, filed Jun. 10, 2015, 8 pages.
  • J.G. Cannon, Chapter Nineteen in Burger's Medicinal Chemistry and Drug Discovery, Fifth Edition, vol. I: Principles and Practice, Wiley-Interscience 1995, pp. 783-802 (Year 1995).
  • Office Action dated Dec. 19, 2017, for Canadian Patent Application No. 2952012, filed on Dec. 12, 2016, 4 pages.
  • Office Action dated Nov. 13, 2017, for New Zealand Patent Application No. 726360, filed Nov. 15, 2016, 4 pages.
  • Office Action dated Oct. 27, 2017, for Australian Patent Application No. 2015274696, filed Nov. 15, 2016, 4 pages.
  • Office Action dated Apr. 3, 2018, for Korean Patent Application No. 10-2017-7000749, filed Sep. 29, 2016, 15 pages (including English translation).
  • Office Action dated Dec. 14, 2017, for Japanese Patent Application No. 2016570274, filed Jun. 10, 2015, 16 pages (with English translation).
  • Office Action dated Dec. 28, 2017, for Eurasian Patent Application No. 201692268,55. filed on Dec. 7, 2016, 8 pages (with English translation).
  • Office Action dated Feb. 6, 2018, for Israel Patent Application No. 248897, filed Nov. 10, 2016, 5 pages (with English translation).
  • Panayotou, G. et al. (Dec. 1992). “Phosphatidyl-Inositol 3-Kinase: A Key Enzyme in Diverse Signaling Processes,” Trends in Cell Biol. 2:358-360.
  • Pinedo, H.M. et al. (2000). “Translational Research: the Role of VEGF in Tumor Angiogenesis,” The Oncologist 5(Suppl. 1):1-2.
  • U.S. Appl. No. 15/680,045, filed Aug. 17, 2017, by Evarts et al.
  • U.S. Appl. No. 16/418,558, filed May 21, 2019, by Kesicki et al. (U.S. Patent Application is not submitted herewith pursuant to the waiver of 37 C.F.R. § 1.98(a)(2)(iii) issued by the Office dated Sep. 21, 2004).
  • U.S. Appl. No. 16/529,213, filed Aug. 1, 2019, by Sadhu et al. (U.S. Patent Application is not submitted herewith pursuant to the waiver of 37 C.F.R. § 1.98(a)(2)(iii) issued by the Office dated Sep. 21, 2004).
  • Khan, F.N. et al. (May 1, 2010, e-pub. Apr. 2, 2010). “2-Chloro-8-methyl-3-[(pyrimidin-4-yl-oxy)meth-yl]quinoline,” Act Crystallogr. Sect. E. Struct. Rep. Online 66(5):O1010-O1010, 10 pages.
  • Canadian Office Action dated Aug. 7, 2018, for Patent Application No. 2,952,012, filed Jun. 10, 2015, 3 pages.
  • Chinese Office Action dated May 3, 2018 for Application No. 201580030997.8, internationally filed Jun. 10, 2015, 16 pages. (including English translation).
  • Eurasian Office Action dated Jul. 5, 2018, for Patent Application No. 201692268, filed Jun. 10, 2015, 4 pages. (including English translation).
  • European Office Action dated Feb. 5, 2018, for Application No. 15 739 038.6, internationally filed Jun. 10, 2015, 4 pages.
  • European Office Action dated Sep. 14, 2018, for Application No. 15 739 038.6, internationally filed Jun. 10, 2015, 4 pages.
  • Japanese Office Action dated Jul. 17, 2018, for Patent Application No. 2016-570309, filed Jun. 10, 2015, 8 pages. (including English translation).
  • Korean Office Action dated Jun. 27, 2018, for Patent Application No. 10-2017-7000749, filed Jun. 10, 2015, 6 pages. (including English translation).
  • New Zealand Notice of Acceptance dated Apr. 12, 2018 for Patent Application No. 726360, internationally filed Jun. 10, 2015, 1 page.
  • New Zealand Office Action dated Apr. 19, 2017, for Patent Application No. 726360, internationally filed Jun. 10, 2015, 4 pages.
  • U.S. Appl. No. 15/993,299, filed May 30, 2018, by Sadhu et al. (U.S. Patent Application is not submitted herewith pursuant to the waiver of 37 C.F.R. § 1.98(a)(2)(iii) issued by the Office dated Sep. 21, 2004).
Patent History
Patent number: 11021467
Type: Grant
Filed: Jun 10, 2015
Date of Patent: Jun 1, 2021
Patent Publication Number: 20150361068
Assignee: Gilead Sciences, Inc. (Foster City, CA)
Inventors: Shaopei Cai (Seattle, WA), Zhimin Du (Belmont, CA), Musong Kim (Bothell, WA), Jennifer A. Loyer-Drew (Seattle, WA), Devan Naduthambi (San Bruno, CA), Leena Patel (Mercer Island, WA), Barton W. Phillips (San Mateo, CA), Gary Phillips (Issaquah, WA), Kirk L. Stevens (Bothell, WA), Jennifer Anne Treiberg (Redmond, WA), Joshua Van Veldhuizen (Seattle, WA), William J. Watkins (Saratoga, CA), Suet Chung Yeung (Redmond, WA)
Primary Examiner: Theodore R. Howell
Application Number: 14/735,244
Classifications
Current U.S. Class: The Additional Hetero Ring Is Six-membered Consisting Of One Nitrogen And Five Carbons (514/266.21)
International Classification: C07D 403/12 (20060101); C07D 401/14 (20060101); C07D 403/14 (20060101); C07D 473/34 (20060101); C07D 487/04 (20060101); C07D 513/04 (20060101); C07D 405/14 (20060101); C07D 413/14 (20060101); C07D 471/04 (20060101);